Language selection

Search

Patent 2663545 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2663545
(54) English Title: NEUROPROTECTIVE PEPTIDE INHIBITORS OF AP-1 SIGNALING AND USES THEREOF
(54) French Title: INHIBITEURS DE SIGNALISATION AP-1 DE PEPTIDE NEUROPROTECTEUR ET UTILISATIONS CORRESPONDANTES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 14/195 (2006.01)
  • A61K 38/10 (2006.01)
  • A61K 38/16 (2006.01)
  • A61P 25/06 (2006.01)
  • A61P 25/08 (2006.01)
  • A61P 25/16 (2006.01)
  • A61P 25/28 (2006.01)
  • C07K 7/08 (2006.01)
  • C07K 14/16 (2006.01)
  • C07K 14/50 (2006.01)
  • C07K 19/00 (2006.01)
  • C12N 15/18 (2006.01)
  • C12N 15/31 (2006.01)
  • C12N 15/49 (2006.01)
(72) Inventors :
  • WATT, PAUL (Australia)
  • MILECH, NADIA (Australia)
  • FEAR, MARK (Australia)
(73) Owners :
  • PHYLOGICA LIMITED (Australia)
(71) Applicants :
  • PHYLOGICA LIMITED (Australia)
(74) Agent: FETHERSTONHAUGH & CO.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2007-01-31
(87) Open to Public Inspection: 2008-03-27
Examination requested: 2012-01-18
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/AU2007/000092
(87) International Publication Number: WO2008/034161
(85) National Entry: 2009-03-13

(30) Application Priority Data:
Application No. Country/Territory Date
60/826,208 United States of America 2006-09-19

Abstracts

English Abstract

The present invention provides AP-1 signaling inhibitory peptides comprising the amino acid sequences of SEQ ID NOS: 28-31, 36-39, 45-48, 54-57 and 62-65, compositions comprising one or a plurality of said peptides, and methods of treatment of neurological disorders comprising the administration of the peptide(s) or compositions.


French Abstract

L'invention concerne des peptides inhibiteurs de signalisation AP-1 qui contiennent des séquences d'acides aminés de SEQ ID NOS: 28-31, 36-39, 45-48, 54-57 et 62-65, des compositions contenant un ou plusieurs de ces peptides, et des procédés de traitement de troubles neurologiques qui consistent à administrer ce(s) peptide(s) ou ces compositions.

Claims

Note: Claims are shown in the official language in which they were submitted.




78

WE CLAIM:


1. A neuroprotective AP-1 signaling inhibitory peptide individually or
collectively
selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
individually or collectively selected from the group consisting of: SEQ ID NO:
28,
SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 36, SEQ ID NO: 37,
SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47,
SEQ ID NO: 48, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57,
SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64 and SEQ ID NO: 65;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
individually or collectively selected from the group consisting of SEQ ID NO:
28, SEQ
ID NO: 29, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 45, SEQ ID NO: 46, SEQ
ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 62 and SEQ ID NO: 63;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions; and

(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.


2. The neuroprotective AP-1 signaling inhibitory peptide of claim 1 wherein
said
peptide is individually or collectively selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
individually or collectively selected from the group consisting of: SEQ ID NO:
28,
SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 54, SEQ ID NO: 55,
SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64
and SEQ ID NO: 65;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence




79

individually or collectively selected from the group consisting of SEQ ID NO:
28, SEQ
ID NO: 29, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 62 and SEQ ID NO: 63;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.


3. The neuroprotective AP-1 signaling inhibitory peptide of claim 1 wherein
said
peptide is a retroinverted peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
37, SEQ ID NO: 39, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 55, SEQ ID NO:
57, SEQ ID NO: 63 and SEQ ID NO: 65;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29, SEQ ID NO: 37, SEQ ID NO: 46, SEQ
ID NO: 55 and SEQ ID NO: 63; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions.


4. The neuroprotective AP-1 signaling inhibitory peptide of claim 1 wherein
said
peptide provides enhanced inhibition of glutamate excitotoxicity in vitro
relative to an
equimolar concentration of the peptide JNK1-1D-TAT (SEQ ID NO: 68).


5. The neuroprotective AP-1 signaling inhibitory peptide of claim 4 wherein
said
peptide significantly inhibits glutamate excitotoxicity at a concentration of
less than
about 2µM.




80

6. The neuroprotective AP-1 signaling inhibitory peptide of claim 4 wherein
said
peptide significantly inhibits glutamate excitotoxicity at a concentration of
less than
about 1µM.


7. The neuroprotective AP-1 signaling inhibitory peptide of claim 4 wherein
said
peptide is selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
37, SEQ ID NO: 39, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO:
48, SEQ ID NO: 55 and SEQ ID NO: 57;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29, SEQ ID NO: 37, SEQ ID NO: 45, SEQ
ID NO: 46 and SEQ ID NO: 55;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.


8. The neuroprotective AP-1 signaling inhibitory peptide of claim 7 wherein
said
peptide is a retroinverted peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
37, SEQ ID NO: 39, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 55 and SEQ ID
NO: 57;

(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected




81

from the group consisting of SEQ ID NO: 29, SEQ ID NO: 37, SEQ ID NO: 46 and
SEQ ID NO: 55; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions.


9. The neuroprotective AP-1 signaling inhibitory peptide of claim 7 wherein
said
peptide is a retroinverted peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
55 and SEQ ID NO: 57;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29 and SEQ ID NO: 55; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions.


10. The neuroprotective AP-1 signaling inhibitory peptide of claim 1 wherein
said
peptide protects neurons from cell death in vivo.


11. The neuroprotective AP-1 signaling inhibitory peptide of claim 10 wherein
said
peptide is a retroinverted peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID
NO:
38, SEQ ID NO: 39, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47 and SEQ ID
NO: 48;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected




82

from the group consisting of SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 45 and
SEQ ID NO: 46;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.


12. The neuroprotective AP-1 signaling inhibitory peptide of claim 10 wherein
said
peptide is selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID
NO:
38 and SEQ ID NO: 39;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 36 and SEQ ID NO: 37;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.


13. The neuroprotective AP-1 signaling inhibitory peptide of claim 10 wherein
said
peptide is a retroinverted peptides selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 37 and SEQ ID NO: 39;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising the sequence
set forth
in SEQ ID NO: 37; and




83

(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% sequence identity thereto and comprising a sequence
that
differs from a sequence set forth in (i) or (ii) by one or more conservative
amino acid
substitutions.


13. The neuroprotective AP-1 signaling inhibitory peptide according to any one
of
claims 1-3, 7-9 or 11-13 wherein the protein transduction domain at (ii)
comprises an
amino acid sequence selected from the group set forth in SEQ ID NOS: 1-25.


14. The neuroprotective AP-1 signaling inhibitory peptide according to any one
of
claims 1-3, 7-9 or 11-13 wherein the protein transduction domain at (ii)
comprises a
TAT basic region peptide comprising a sequence set forth in any one of SEQ ID
NOS:
1-16.


15. The neuroprotective AP-1 signaling inhibitory peptide according to any one
of
claims 1-3, 7-9 or 11-13 wherein the protein transduction domain and the
peptide at (ii)
are separated by a linker.


16. The neuroprotective AP-1 signaling inhibitory peptide of claim 15 wherein
the
linker comprises from 1 to about 6 glycine residues or other amino acids of
low
immunogenicity.


17. The neuroprotective AP-1 signaling inhibitory peptide of claim 16 wherein
the
linker comprises a glycine residue.


18. A neuroprotective composition comprising (i) an amount of a
neuroprotective
AP-1 signaling inhibitory peptides according to any one of claims 1 to 17
sufficient to
reduce, delay or prevent neuronal apoptosis and/or necrosis in an animal; and
(ii) a
suitable carrier or excipient for application to the central nervous system of
the animal.




84

19. The neuroprotective composition of claim 18 comprising a plurality of said

neuroprotective AP-1 signaling inhibitory peptides.


20. The neuroprotective composition of claim 18 further comprising JNK
inhibitory
peptide JNK1-1D-TAT (SEQ ID NO: 68).


21. The neuroprotective composition of claim 18 further comprising a
composition
of matter having synergistic activity with respect to neuron repair.


22. A method for preventing or delaying neuronal cell death in a subject
comprising
administering an AP-1 signaling inhibitory peptide according to any one of
claims 1-17
or a composition comprising said peptide to a subject in need of treatment.


23. The method of claim 22 wherein the subject has developed or suffers from a

neuronal condition involving glutamate and/or NMDA excitotoxicity and/or
ischemia
and/or wounding.


24. The method of claim 23 wherein the subject has suffered from or is likely
to
suffer from migraine, cerebral ischemia, traumatic brain injury, epilepsy,
Parkinson's
Disease, Alzheimer's Disease and Amyotrophic Lateral Sclerosis (ALS).


25. The method according to claim 24 wherein the subject has suffered local or

global cerebral ischemia or traumatic brain injury or acute cortical injury.


26. The method according to claim 25 wherein the local or global cerebral
ischemia
or traumatic brain injury is a recent event.


27. The method according to claim 22 wherein the peptide or composition is
administered to a subject by producing a recombinant cell expressing said
peptide and
administering a composition comprising said cell to the subject.




85

28. The method of claim 27 wherein the cell is a stem cell.


29. The method of claim 22 wherein the peptide or composition is administered
to a
subject by particle bombardment of nucleic acid encoding the peptide under
conditions
sufficient for transcription and translation of said nucleic acid to occur.


30. The method of claim 22 comprising repeated administration of the peptide
or
composition.


31. The method of claim 22 comprising:
(i) identifying a subject suffering from or developing a neuronal condition
involving glutamate and/or NMDA excitotoxicity and/or ischemia and/or
wounding;
(ii) obtaining an AP-1 signaling inhibitory peptide according to any one of
claims 1-
17 or a composition comprising said peptide; and
(iii) administering said peptide or composition to said subject.

32. The method of claim 22 comprising:
(i) identifying a subject suffering from or developing a neuronal condition
involving glutamate and/or NMDA excitotoxicity and/or ischemia and/or
wounding; and
(ii) recommending administration of an AP-1 signaling inhibitory peptide
according
to any one of claims 1-17 or a composition comprising said peptide.


33. The method of claim 22 comprising administering or recommending
administration of an AP-1 signaling inhibitory peptide according to any one of
claims
1-17 or a composition comprising said peptide to a subject previously
identified as
suffering from a neuronal condition involving glutamate and/or NMDA
excitotoxicity
and/or ischemia and/or wounding.


34. Use of an amount of an AP-1 signaling inhibitory peptide according to any
one
of claims 1-17 sufficient to inhibit or delay neuronal cell death in the
manufacture of a
medicament for the treatment of a neuronal condition involving glutamate
and/or
NMDA excitotoxicity and/or ischemia and/or wounding.


Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-1-
Neuroprotective peptide inhibitors of AP-1 signaling and uses thereof
Related application data
The present invention claims priority from US Patent Application No.
60/826,208 filed
September 19, 2006 which is herein incorporated by reference in its entirety.

Field of the invention
The present invention relates generally to peptides having neuroprotective
properties by
virtue of inhibiting AP-1 signaling in mammalian cells. Also provided are
methods for
1o the diagnosis and treatment of aberrant neuronal function by virtue of
inhibiting,
delaying or preventing AP-1 signaling neuronal disorders stroke using the
peptides of
the invention.

Background of the invention
1. General information
This specification contains nucleotide and amino acid sequence information
prepared
using Patentln Version 3.3, presented herein after the claims. Each nucleotide
sequence is identified in the sequence listing by the numeric indicator <210>
followed
by the sequence identifier (e.g. <210>1, <210>2, <210>3, etc). The length and
type of
sequence (DNA, protein (PRT), etc), and source organism for each nucleotide
sequence, are indicated by information provided in the numeric indicator
fields <211>,
<212> and <213>, respectively. Nucleotide sequences referred to in the
specification
are defined by the term "SEQ ID NO:", followed by the sequence identifier (eg.
SEQ
ID NO: 1 refers to the sequence in the sequence listing designated as <400>1).

The designation of nucleotide residues referred to herein are those
recommended by the
IUPAC-IUB Biochemical Nomenclature Commission, wherein A represents Adenine,
C represents Cytosine, G represents Guanine, T represents thymine, Y
represents a
pyrimidine residue, R represents a purine residue, M represents Adenine or
Cytosine, K
3o represents Guanine or Thymine, S represents Guanine or Cytosine, W
represents
Adenine or Thymine, H represents a nucleotide other than Guanine, B represents
a


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-2-
nucleotide other than Adenine, V represents a nucleotide other than Thymine, D
represents a nucleotide other than Cytosine and N represents any nucleotide
residue.

As used herein the term "derived from" shall be taken to indicate that a
specified
integer may be obtained from a particular source albeit not necessarily
directly from
that source.

Throughout this specification, unless the context requires otherwise, the word
"comprise", or variations such as "comprises" or "comprising", will be
understood to
io imply the inclusion of a stated step or element or integer or group of
steps or elements
or integers but not the exclusion of any other step or element or integer or
group of
elements or integers.

Throughout this specification, unless specifically stated otherwise or the
context
requires otherwise, reference to a single step, composition of matter, group
of steps or
group of compositions of matter shall be taken to encompass one and a
plurality (i.e.
one or more) of those steps, compositions of matter, groups of steps or group
of
compositions of matter.

2o Each embodiment described herein is to be applied mutatis mutandis to each
and every
other embodiment unless specifically stated otherwise.

Those skilled in the art will appreciate that the invention described herein
is susceptible
to variations and modifications other than those specifically described. It is
to be
understood that the invention includes all such variations and modifications.
The
invention also includes all of the steps, features, compositions and compounds
referred
to or indicated in this specification, individually or collectively, and any
and all
combinations or any two or more of said steps or features.

3o The present invention is not to be limited in scope by the specific
embodiments
described herein, which are intended for the purpose of exemplification only.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-3-
Functionally-equivalent products, compositions and methods are clearly within
the
scope of the invention, as described herein.

The present invention is performed without undue experimentation using, unless
otherwise indicated, conventional techniques of molecular biology,
microbiology,
virology, recombinant DNA technology, peptide synthesis in solution, solid
phase
peptide synthesis, and immunology. Such procedures are described, for example,
in the
following texts:
1. Sambrook, Fritsch & Maniatis, , whole of Vols I, II, and III;
io 2. DNA Cloning: A Practical Approach, Vols. I and II (D. N. Glover, ed.,
1985),
IRL Press, Oxford, whole of text;
3. Oligonucleotide Synthesis: A Practical Approach (M. J. Gait, ed., 1984) IRL
Press, Oxford, whole of text, and particularly the papers therein by Gait, pp1-
22;
Atkinson et al., pp35-81; Sproat et al., pp 83-115; and Wu et al., pp 135-151;
4. Nucleic Acid Hybridization: A Practical Approach (B. D. Hames & S. J.
Higgins, eds., 1985) IRL Press, Oxford, whole of text;
5. Animal Cell Culture: Practical Approach, Third Edition (John R.W. Masters,
ed., 2000), ISBN 0199637970, whole of text;
6. Immobilized Cells and Enzymes: A Practical Approach (1986) IRL Press,
Oxford, whole of text;
7. Perbal, B., A Practical Guide to Molecular Cloning (1984);
8. Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press,
Inc.), whole of series;
9. J.F. Ramalho Ortigao, "The Chemistry of Peptide Synthesis" In: Knowledge
database of Access to Virtual Laboratory website (Interactiva, Germany);
10. Sakakibara, D., Teichman, J., Lien, E. Land Fenichel, R.L. (1976).
Biochem.
Biophys. Res. Commun. 73 336-342
11. Merrifield, R.B. (1963). J. Am. Chem. Soc. 85, 2149-2154.
12. Barany, G. and Merrifield, R.B. (1979) in The Peptides (Gross, E. and
Meienhofer, J. eds.), vol. 2, pp. 1-284, Academic Press, New York.
13. Wiinsch, E., ed. (1974) Synthese von Peptiden in Houben-Weyls Metoden der


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-4-
Organischen Chemie (Muler, E., ed.), vol. 15, 4th edn., Parts 1 and 2, Thieme,
Stuttgart.
14. Bodanszky, M. (1984) Principles of Peptide Synthesis, Springer-Verlag,
Heidelberg.
15. Bodanszky, M. & Bodanszky, A. (1984) The Practice of Peptide Synthesis,
Springer-Verlag, Heidelberg.
16. Bodanszky, M. (1985) Int. J. Peptide Protein Res. 25, 449-474.
17. Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C.
Blackwell, eds., 1986, Blackwell Scientific Publications).
io 18. McPherson et al., In: PCR A Practical Approach., IRL Press, Oxford
University
Press, Oxford, United Kingdom, 1991.
19. Methods in Yeast Genetics: A Cold Spring Harbor Laboratory Course Manual
(D. Burke et al., eds) Cold Spring Harbor Press, New York, 2000 (see whole of
text).
20. Guide to Yeast Genetics and Molecular Biology. In: Methods in Enzymology
Series, Vol. 194 (C. Guthrie and G.R. Fink eds) Academic Press, London, 1991
2000 (see whole of text).

2. Description of the related art.
Peptide therapeutics
As a response to the increasing demand for new lead compounds and new target
identification and validation reagents, the pharmaceutical industry has
increased its
screening of various sources for new lead compounds having a unique activity
or
specificity in therapeutic applications, such as, for example, in the
treatment of
neoplastic disorders, infection, modulating immunity, autoimmunity, fertility,
etc.

It is known that proteins bind to other proteins, antigens, antibodies,
nucleic acids, and
carbohydrates. Such binding enables the protein to effect changes in a wide
variety of
biological processes in all living organisms. As a consequence, proteins
represent an
important source of natural modulators of phenotype. Accordingly, peptides
that
modulate the binding activity of a protein represent attractive lead compounds
(drug


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-5-
candidates) in primary or secondary drug screening. For example, the formation
of a
target biological interaction that has a deleterious effect (eg. replication
of a pathogen
or of a cancer cell), can be assayed to identify lead compounds that
antagonize the
biological interaction.

Antibodies represent the fastest growing class of approved drugs in this area,
however
they require complex and expensive synthesis and are difficult to deliver via
non-
injectable routes. In contrast, large peptides can often be made synthetically
and are
increasingly being delivered by buccal, intranasal or intranasal routes as
alternatives to
1o injection. Furthermore, intracellular delivery of peptides is also now
possible in vivo
using protein transduction domains. These advances make peptide-based
therapeutics
an attractive alternative to antibody-based therapeutics.

Existing drawbacks associated with peptide-based therapeutics include their
low
affinity, high turnover in vivo and difficulties in their isolation compared
to small
molecules. For example, peptides that target protein interaction interfaces
which may
be large and relatively featureless are generally more difficult to produce
and isolate
when compared to small molecule inhibitors of enzyme-active sites that
generally form
small complex pockets. Accordingly, it is not facile to identify peptides that
address
these problems.

For example, random peptide (synthetic mimetic or mimotope) libraries can be
produced using short random oligonucleotides produced by synthetic
combinatorial
chemistry, cloned into an appropriate vehicle for expression, and the encoded
peptide
screened using one of a variety of approaches. However, the ability to isolate
active
peptides from random fragment libraries can be highly variable with low
affinity
interactions occurring between many of the peptide-binding partners and very
low hit-
rates for biologically active peptides. Moreover, the expressed peptides often
show
little or none of the secondary or tertiary structure required for efficient
binding
3o activity, and/or are unstable. This is not surprising, considering that
biological
molecules appear to recognize shape and charge rather than primary sequence
(Yang


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-6-
and Honig J. Mol. Biol 301(3), 691-711 2000) and that such random peptides are
generally too small to comprise a protein domain or to form the secondary
structure of
a protein domain. Moreover even the largest peptide libraries to have been
produced
do not contain sufficient complexities to exhaustively cover all of the
possible
combinations of the 20 amino acids, for peptides of more than approximately a
dozen
residues. The relatively unstructured `linear' nature of many artificial
peptides derived
from random amino acid sequences also leads to their more rapid degradation
and
clearance following administration to a subject in vivo, thereby reducing
their appeal as
therapeutic agents.
In contrast, natural protein folds or subdomains are understood in the art to
mean
independently folding peptide structures (e.g., a 19-residue fragment from the
C-loop
of the fourth epidermal growth factor-like domain of thrombomodulin as been
described by Alder et al, J. Biol. Chem., 270: 23366-23372, 1995). These
constrained
structures provide thermodynamic advantages to bind other protein surfaces
through
limiting the entropic cost of binding. Moreover, structured folds can be less
susceptible
to proteolysis than unstructured linear peptides, increasing their biological
stability.

To enhance the probability of obtaining useful bioactive peptides or proteins
from
2o random peptide libraries, peptides have previously been constrained within
scaffold
structures, eg., thioredoxin (Trx) loop (Blum et al. Proc. Natl. Acad. Sci.
USA, 97,
2241-2246, 2000) or catalytically inactive staphylococcal nuclease (Norman et
al,
Science, 285, 591-595, 1999), to enhance their stability. Constraint of
peptides within
such structures has been shown, in some cases, to enhance the affinity of the
interaction
between the expressed peptides and its target, presumably by limiting the
degrees of
conformational freedom of the peptide, and thereby minimizing the entropic
cost of
binding.

Recently, peptide mimotopes of less than about 50 amino acids in length have
been
3o described that are capable of forming protein domains by virtue of assuming
conformations sufficient for binding to a target protein or target nucleic
acid


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-7-
("PhylomerTM peptides", Phylogica, Perth, western Australia, Australia) e.g.,
International Patent Application No. PCT/AUOO/00414 and US Patent Publication
No.
2003-0215846 Al. Such PhylomerTM . peptides show promise in overcoming the
existing drawbacks associated with peptide therapeutics. The conformation(s)
of such
PhylomerTm peptides is a product of secondary and/or tertiary structural
features and,
by virtue of the peptide binding to its target protein or protein interaction
interface is
compatible with, albeit not necessarily iterative of, the target protein(s) or
target protein
interaction interface. Such secondary structural features may suggest that
PhylomerTM
peptides, on average, have higher substrate affinities and longer half-lives
than more
1o conventional random peptides. On the other hand, PhylomerTM peptides may
also
provide production and delivery advantages compared to antibody-based
therapies by
virtue of their small size. Additionally, because PhylomerTM peptides are
derived from
libraries comprising mixtures of small genome fragments from evolutionarily-
diverse
bacteria and eukaryotes having small albeit well-characterized genomes, they
can be
screened in silico to select against those peptides sequences that are likely,
because of
their known strucure or function, to produce adverse reactions in recipient
mammals,
including humans. Notwithstanding the need for empirical testing of
therapeutic
products, this "safety" feature of PhylomerTm peptides provides a significant
potential
advantage over peptides derived from mammals, including antibodies.

Neuronal disorders involving neuronal cell death
Neuronal disorders such as migraine, stroke, traumatic brain injury, epilepsy
and
neurodegenerative disorders including Huntington's Disease (HD), Parkinson's
Disease
(PD), Alzheimer's Disease (AD) and Amyotrophic Lateral Sclerosis (ALS) are
major
causes of morbidity and disability arising from long term brain injury. These
effects
generally involve apoptosis and/or necrosis of neurons, possibly involving
diverse
pathways including oxidative stress.

As used herein, the term "stroke" includes any ischemic disorder e.g., a
peripheral
vascular disorder, a venous thrombosis, a pulmonary embolus, a myocardial
infarction,
a transient ischemic attack, lung ischemia, unstable angina, a reversible
ischemic


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-8-
neurological deficit, adjunct thromolytic activity, excessive clotting
conditions,
reperfusion injury, sickle cell anemia, a stroke disorder or an iatrogenically
induced
ischemic-period such as angioplasty, or cerebral ischemia.

Glutamate excitotoxicity
Increased extracellular levels of the neurotransmitter glutamate cause
neuronal cell
death via excitotoxicity. An accumulation of extracellular glutamate over-
stimulates
NMDA and AMPA receptors resulting in an influx of extracellular calcium and
sodium
ions and the release of bound calcium from intracellular stores. The increase
in
1o intracellular calcium initiates a range of cell damaging events involving
phospholipases, proteases, phosphatases, kinases and nitric oxide synthase, as
well as
the activation of the pro-apoptotic transcription factor c-Jun.

Involvement of the AP-1 signaling pathway in neuronal function
Various types of evidence indicate that c-Jun N-Terminal Kinase (JNK or SAPK)
is
involved in neuronal cell death during or following ischemia, via activation
of c-Jun (a
component of the AP-1 complex) in an analogous way to the known activation of
this
stress kinase response in other forms of ischemia such as coronary heart
disease or in
organ or blood vessel reperfusion injury.

Components of the AP-1 pathway associate with scaffold proteins that modulate
their
activities and cellular localization. JNK activity is controlled by a cascade
of protein
kinases and by protein phosphatases, including dual-specificity MAPK
phosphatases.
For example, the JNK-interacting protein-1 (JIP-1) scaffold protein
specifically binds
JNK, MAPK kinase 4 (MKK4) and MAPK kinase 7 (MKK7), and members of the
mixed lineage kinase (MLK) family, and regulates JNK activation in neurons.
Distinct
regions within the N termini of MKK7 and the MLK family member dual leucine
zipper kinase (DLK) mediate their binding to JIP-1. JNK binds to c-Jun, and
this
appears to be required for efficient c-Jun phosphorylation.

Several members of the death-related AP-1 pathway acting upstream of JNK have
been


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-9-
defined. The most distal of these are the Rho small GTPase family members Racl
and
Cdc42. Over expression of constitutively active forms of Rac 1(i. e., Rac l V
12) and
Cdc42 (i.e., Cdc42Vl2) leads to activation of the AP-1 pathway and to death of
Jurkat
T lymphocytes, PC12 cells, and sympathetic neurons. Conversely, over
expression of
dominant-negative mutants of Cdc42 (i.e., Cdc42N17) and Racl (i.e., Rac1N17)
in
sympathetic neurons prevents elevation of c-Jun and death evoked by nerve
growth
factor (NGF) withdrawal (Bazenet et al., Proc. Natl. Acad. Sci. USA 95, 3984-
3989,
1998; Chuang et al., Mol. Biol. Cell 8, 1687-1698, 1997). Over expression of
the
dominant negative mutant RaclN17 also reverses the induction of death by
Cdc42V12,
io whereas Cdc42N 17 has no effect on Rac 1 V 12-induced death, suggesting
that Cdc42
lies upstream of Racl (Bazenet et al., Proc. Natl. Acad. Sci. USA 95, 3984-
3989, 1998).
Similar approaches have indicated that mitogen-activated protein kinase
kinases 4 and
7 (MKK4 and MKK7) lie downstream of Cdc42 and Rac1 and directly upstream of
the
JNKs (Foltz et al., J Biol. Chem. 273, 9344-9351, 1998; Holland et al., J.
Biol. Claem.
~72, 24994-24998, 1997; Mazars et al., Oncogene 19, 1277-1287, 2000; Vacratsis
et
al., J. Biol. Chem. 275, 27893-27900, 2000; Xia et al., Science 270, 1326-
1331, 1995;
Yamauchi et al., J. Biol. Chem. 274, 1957-1965, 1999). Studies using
constitutively
active and dominant-negative constructs have also implicated apoptosis signal-
regulating kinase 1(ASK1) as an additional participant in the pathway that
lies between
20= Cdc42 and the downstream MKKs and JNKs (Kanamoto et al., Mol. Cell. Biol.
20,
196-204, 2000).

MLKs have been shown to function as MKK kinases and lead to activation of JNKs
via
activation of MKKs (Bock et al., J. Biol. Chem. 275, 14231-1424, 2000; Cuenda
et al.,
Biochem. J. 333, 11-159, 1998; Hirai et al., J. Biol. Claem. 272, 15167-15173,
1997;
Merritt et al., J. Biol. Chem. 274, 10195-10202, ;1999; Rana et al., J. Biol.
Chem. 271,
19025-19028, 1996; Tibbles et al., EMBO J. 15, 7026-7035, 1996; Vacratsis et
al., J.
Biol. Chem. 275, 27893-27900, 2000). Members of the family include MLK1, MLK2
(also called MST), MLK3 (also called SPRK or PTK1), dual leucine zipper kinase
(DLK; also called MUK or ZPK), and leucine zipper-bearing kinase (LZK).
Constitutively active mutants of Racl and Cdc42 have been found to bind to and
to


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-10-
modulate the activities of MLK2 and -3, and co-expression of MLK3 and
activated
Cdc421eads to enhanced MLK3 activation.

In animal models of ischemia or migraine, stroke, apoptotic neurons have
enhanced
phosphorylation of the transcription factor c-Jun by JNK. Additionally,
neuronal c-Jun
levels are elevated in response to trophic factor withdrawal, and dominant-
negative
forms of this transcription factor are at least partially-protective against
neuronal cell
death evoked by selective activation of JNKs (Eilers et al., J. Neurosci. 18,
1713-1724,
1998; Ham et al., Neuron 14, 927-939).
The transcriptional activating activity of c-Jun is regulated at the post-
translational
level by its phosphorylation by JNK (SAPK) at two residues within the amino-
terminal
trans-activation domain, serines 63 and 73, in response to a variety of
cellular stresses.
Phosphorylation of these two residues is critical for the transcriptional
activating
activity of c-Jun, since mutation of them markedly decreases this activity.
JNKs
(SAPKs) readily phosphorylate c-Jun at Ser 63/73, and at a rate that is about
10 times
faster than ERK-1 and ERK-2. The JNKs (SAPKs) account for the majority of c-
Jun
trans-activation domain (Ser 63/73) kinase activity after reperfusion,
suggesting that
they trigger part of the kidney's very early genetic response to ischemia by
enhancing
the transcriptional activating activity of c-Jun. Since induction of c-Jun is
auto-
regulated, it is likely that activation of the JNKs (SAPKs) is, at least in
part, responsible
for the induction of c-Jun following myocardial or renal ischemia.

The role of JNKs (SAPKs) in the control of gene expression during and/or
following
ischemia extends well beyond the regulation of c-Jun by JNK. It is known that
AP-1
comprises complexes of c-Jun with parters such as c-Fos or ATF-2 (a member of
the
CREB family). When complexed with c-Fos, the dimer is targeted to promoters,
such
as that of the collagenase gene, containing canonical AP-1 elements. When
complexed
with ATF-2, however, the dimer appears to prefer CRE sequences, and AP-1
variants
such as that contained in the c-Jun promoter which controls induction of c-Jun
in
response to a variety of stimuli. After ischemia and reperfusion, ATF-2 and c-
Jun are


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-11-
targeted as a heterodimer to both ATF/CRE motifs and the Jun2 TRE within the c-
Jun
promoter. This suggests that, following reperfusion of ischemic tissue, the
JNKs
(SAPKs) target ATF-2/c-Jun heterodimers to various promoters, including the c-
Jun
promoter, and enhance transcriptional activating activity of both components
of the c-
Jun/ATF-2 dimer. This may provide a potent mechanism for the induction of a
large
number of genes regulated by promoters containing ATF/CRE sites or AP- 1
variants to
which the heterodimer binds.

Dimerization of c-Jun also leads to apoptosis in neurons in response to
ischemia (Tong
1o et al., J. Neurochem 71, 447-459, 1998; Ham et al., Biochem. Pharmacol. 60,
1015-
1021, 2000).

A homodimer of c-Jun is also known to activate the c-Jun transcription factor
via
binding to the transcriptional regulatory element (TRE) in the c-Jun promoter.

As used herein unless specifically stated otherwise or the context requires
otherwise,
the term "c-Jun dimerization" shall be taken to include homo-dimerization of c-
Jun
monomers and the partnering of c-Jun with another peptide or polypeptide e.g.,
JNK, c-
Fos, ATF-2. Similarly, unless specifically stated otherwise or the context
requires
otherwise, the term "c-Jun dimer" shall be taken to include homo-dimer of c-
Jun
monomers and a heterodimer of c-Jun with another peptide or polypeptide e.g.,
c-Fos,
ATF-2, including transient complexes such as those between the JNK kinase and
its
substrate c-Jun.

Treatment of neuronal cell death
Currently, there is no effective clinical agent that inhibits the delayed
neuronal cell
death associated with such neuronal dysfunction. For example, drugs such as
Activase
(genetically engineered tissue plasminogen activator; Genentech), Abciximab (a
platelet inhibitor; Centocor), and Ancrod (fibrinogenolytic) have had limited
success,
3o even if administered soon after the stroke occurs. These agents offer no
clinical benefit
if administered later than the period immediately following the stroke and
unfortunately


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-12-
many patients present to a hospital after this window of opportunity. Even
alternative
approaches that target glutamate receptors to prevent glutamate excitotoxicity
causing
neuronal damage have shown no significant or consistent improvements in
patient
outcome, most likely due to the need to target these events early.

Summary of the invention
The present invention is based upon the identification by the inventors of
PhylomerTm
peptides that inhibit AP-1 signaling as determined by binding to human c-Jun
in yeast
cells, and AP-1 regulated transcription in mammalian cells. The peptides were
io identified using a reverse hybrid screening technology that employed dual
counter
selection using the cytotoxic compounds cycloheximide and 5-fluoro orotic acid
(5-
FOA), in which only cells in which an interaction between JUN1 and JUNZ is
disrupted could be rescued (Example 1). PhylomerTm peptides that rescued yeast
cells
in primary reverse hybrid screens were then expressed in mammalian cells
expressing
luciferase under operable control of AP-1 enhancer elements, to confirm their
ability to
inhibit AP-1 regulated transcription (Example 2).

By virtue of their activity in yeast cells in preventing JUN1/JUNZ
dimerization, the
identified PhylomerTM peptides are candidate AP-1 signaling inhibitory
peptides that
inhibit AP-1 signaling by direct inhibition of c-Jun homodimerization and/or
heterodimerization. Such a mechanism of action is entirely consistent with the
ability
of the PhylomerTM peptides to also prevent expression of a luciferase reporter
gene in
mammalian cells.

The AP-1 binding Phylomerm peptides can also be validated by other related
methodologies, such as forward two hybrid screens using c-Jun as a bait.
Peptide
inhibitors of c-Jun/dependent autoactivation in two hybrid assays can be
captured using
counterselection approaches such as those described above. Similarly
inhibitors of c-
Jun to APl binding sites can be validated through standard one hybrid assays
using this
promoter element. A subset of such c-Jun binding, or DNA binding peptides
might be
expected to also inhibit AP 1 signalling.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-13-
It is also possible that certain PhylomerTM peptides inhibit AP-1 signaling by
indirect
means e.g., involving factors upstream of c-Jun that are conserved between
yeasts and
mammals. For example, yeast cells possess a stress-responsive MAPK (SAPK)
cascade; a multistep phosphorelay system; and AP-1-like transcription factor
(Yap 1)
that govern the response of yeasts to oxidative stress (Ikner et al., Mutation
Res. 569,
13-27, 2005), and which may be involved in regulating the apoptotic response
to
cytotoxic compounds used in the reverse hybrid screens. The yeast MAPK (SAPK)
1o cascade involves signaling from a complex comprising yeast homologs of
human
Cdc42 and Pakl (i.e., Cdc42 and Ste20, respectively) to the MAPKKK Stel 1,
which
regulates the MAPKK Pbs2 and, in turn, the MAPK Hogl to regulate gene
expression,
membrane transport, cell cycle progression, etc. The yeast phosphorelay system
appears to converge on Pbs2 MAPKK of the Hogl SAPK cascade and is initiated by
the transmembrane protein Shol which activates Pbs2 through the MAPKKK Ste11
of
the Hogl SAPK cascade. The AP-1-like transcription factor (Yapl) appears to
serve as
an oxidative stress sensor that directly regulates transcription albeit
independently of
the SAPK pathway. Without being bound by any theory or mode of action, the
present
inventors reason that a PhylomerTM peptide identified in a counter selection
screen such
2o as a reverse hybrid screening of yeast cells may rescue yeast cells from an
event
upstream of Hogl in yeast that would otherwise lead to activation of these
stress
responses (including cell-cycle modulation) leading to cell death. If the same
PhylomerTM peptide also recognizes a homologous mammalian AP-1 pathway
component upstream of c-Jun and/or JNK, inhibition of that component would
also
explain the observed reduction in AP-1 mediated activation of luciferase
reporter gene
expression observed in mammalian cells.

Accordingly, the identified PhylomerTm peptides from yeast reverse hybrid
screens not
to be limited by their ability to inhibit c-Jun dimerization, and are
designated herein as
"AP-1 inhibitors" or "AP-1 complex formation inhibitors" or "AP-1 signaling
inhibitors" or similar term. It is to be understood that such terminology
includes the


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-14-
direct c-Jun dimerization and/or upstream indirect effects e.g., acting on
phosphorylation of Cdc42, Pakl or Racl, or dimerization of Cdc42, Pakl or Racl
in
mammalian cells. Preferred AP-1 signaling inhibitory peptides will inhibit
later steps
in the AP-1 signaling pathway e.g., c-Jun dimerization, to thereby provide
greater
specificity than, for example, a JNK inhibitory peptide.

It is also to be understood that the term "c-Jun dimerization" includes c-Jun
self-
dimerization or homodimerization, and heterodimerization between c-Jun and
another
protein e.g., ATF-2, c-Fos or JNK and preferably between c-Jun and ATF-2 or
between
io c-Jun and c-Fos (i.e., a c-Jun heterodimer) or an analog of said isolated
peptide or
protein domain.

The present inventors have shown herein that five AP-1 signaling inhibitory
peptides,
designated PYC19, PYC35, PYC36, PYC38/39 and PYC41, are also neuroprotective
in
in vitro and in vivo models of neurological damage in humans. The sequences of
these
peptides are set forth in Table 1 herein and the accompanying Sequence
Listing. In
particular, the PhylomerTM peptides are neuroprotective following glutamate
and/or
NMDA excitotoxicity in primary cortical neuronal cultures, establishing their
relevance
to therapy of disorders such as migraine, stroke, traumatic brain injury,
epilepsy and
2o neurodegenerative disorders including Parkinson's Disease (PD), Alzheimer's
Disease
(AD) and Amyotrophic Lateral Sclerosis (ALS). The inventors have also
demonstrated
that AP-1 signaling inhibitory PhylomerTM peptides, in particular PYC35 and/or
PYC36, protect cultured cortical neurons in an in vitro model of cerebral
ischemia i.e.,
Oxygen Glucose Deprivation (OGD). The data presented herein also demonstrate
that
AP-1 signaling inhibitory PhylomerTm peptides, in particular PYC35 and/or
PYC36,
are neuroprotective in vivo, in a head injury model of global cerebral
ischemia, as
determined by MAP2 immunoreactivity (a marker of neuron loss in brain tissue)
following administration of peptides. In particular, peptide PYC35 provides
significant neuroprotection following acute cortical injury of rat brain
tissue in situ,
including: (i) reduced loss of neurons as determined by MAP2 immunoreactivity;
(ii)
reduced astrogliosis as determined by glial fibrillary acidic protein
immunoreactivity;


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-15-
and (iii) reduced microglial activation as determined by ferritin
immunoreactivity.
These data are consistent with a neuroprotective function and therapeutic
potential in
the treatments of head injury and/or ischemia.

Accordingly, the present invention provides a neuroprotective AP-1 signaling
inhibitory peptide individually or collectively selected from the group
consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
individually or collectively selected from the group consisting of: SEQ ID NO:
28,
SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 36, SEQ ID NO: 37,
lo SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47,
SEQ ID NO: 48, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57,
SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64 and SEQ ID NO: 65;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
individually or collectively selected from the group consisting of SEQ ID NO:
28, SEQ
ID NO: 29, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 45, SEQ ID NO: 46, SEQ
ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 62 and SEQ ID NO: 63;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.

By "individually" is meant that the invention encompasses the recited
neuroprotective
peptides or groups of neuroprotective peptides separately, and that,
notwithstanding
that individual peptides or groups of peptides may not be separately listed
herein the
accompanying claims may define such peptides or groups of peptides separately
and
divisibly from each other.



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-16-
By "collectively" is meant that the invention encompasses any number or
combination
of the recited neuroprotective peptides or groups of neuroprotective peptides,
and that,
notwithstanding that such numbers or combinations of peptides or groups of
peptides
may not be specifically listed herein the accompanying claims may define such
combinations or sub-combinations separately and divisibly from any other
combination
of peptides or groups of peptides.

In another example, the neuroprotective AP-1 signaling inhibitory peptide is
individually or collectively selected from the group consisting of:
1o (i) a neuroprotective AP-1 signaling inhibitory peptide comprising a
sequence
individually or collectively selected from the group consisting of: SEQ ID NO:
28,
SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 54, SEQ ID NO: 55,
SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64
and SEQ ID NO: 65;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
individually or collectively selected from the group consisting of SEQ ID NO:
28, SEQ
ID NO: 29, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 62 and SEQ ID NO: 63;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
2o having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.
In another example, the neuroprotective AP-1 signaling inhibitory peptide is a
retroinverted peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
3o 37, SEQ ID NO: 39, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 55, SEQ ID NO:
57, SEQ ID NO: 63 and SEQ ID NO: 65;


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-17-
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29, SEQ ID NO: 37, SEQ ID NO: 46, SEQ
ID NO: 55 and SEQ ID NO: 63; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions.

1o In another example, the neuroprotective AP-1 signaling inhibitory peptide
provides for
greater inhibition of glutamate excitotoxicity than an equimolar concentration
of the
peptide JNK1-ID-TAT (SEQ ID NO: 68) which inhibits JNK, and preferably
significantly inhibits glutamate excitotoxicity at a concentration of less
than about 1 gM
or 2 M. In accordance with this example, a preferred neuroprotective AP-1
signaling
inhibitory peptide is selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
37, SEQ ID NO: 39, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO:
48, SEQ ID NO: 55 and SEQ ID NO: 57;
2o (ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a
fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29, SEQ ID NO: 37, SEQ ID NO: 45, SEQ
ID NO: 46 and SEQ ID NO: 55;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions; and
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-18-
Preferably, the neuroprotective AP-1 signaling inhibitory peptide is a
retroinverted
peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
37, SEQ ID NO: 39, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 55 and SEQ ID
NO: 57;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29, SEQ ID NO: 37, SEQ ID NO: 46 and
1o SEQ ID NO: 55; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions.

Alternatively, the neuroprotective AP-1 signaling inhibitory peptide is a
retroinverted
peptide selected from the group consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID
NO:
2o 55 and SEQ ID NO: 57;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 29 and SEQ ID NO: 55; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions.

In yet another example, the neuroprotective AP-1 signaling inhibitory peptide
protects
neurons from cell death in vivo. In accordance with this example, a preferred


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-19-
neuroprotective AP-1 signaling inhibitory peptide is selected from the group
consisting
of:

(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID
NO:
38, SEQ ID NO: 39, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47 and SEQ ID
NO: 48;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 45 and
1o SEQ ID NO: 46;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions; and

(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.

Preferred neuroprotective AP-1 signaling inhibitory peptides having
neuroprotective
activity in vivo are selected from the group consisting of:

(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID
NO:
38 and SEQ ID NO: 39;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
between a protein transduction domain and a peptide comprising a sequence
selected
from the group consisting of SEQ ID NO: 36 and SEQ ID NO: 37;
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
3o acid substitutions; and


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-20-
(iv) a retroinverted analog of (i) or (ii) or (iii) or (iv) comprising one or
more D-
amino acids.

More preferably, neuroprotective AP-1 signaling inhibitory peptides having
neuroprotective activity in vivo are retroinverted peptides selected from the
group
consisting of:
(i) a neuroprotective AP-1 signaling inhibitory peptide comprising a sequence
selected from the group consisting of: SEQ ID NO: 37 and SEQ ID NO: 39;
(ii) a neuroprotective AP-1 signaling inhibitory peptide comprising a fusion
io between a protein transduction domain and a peptide comprising the sequence
set forth
in SEQ ID NO: 37; and
(iii) a neuroprotective AP-1 signaling inhibitory peptide that is a variant of
(i) or (ii)
having at least about 90% or 95% sequence identity thereto and comprising a
sequence
that differs from a sequence set forth in (i) or (ii) by one or more
conservative amino
acid substitutions.

In each of the foregoing embodiments, a preferred protein transduction domain
will
comprise an amino acid sequence selected from the group set forth in SEQ ID
NOS: 1-
25, more preferably a peptide transduction domain comprising a sequence
selected
from the group set forth in SEQ ID NOS: 1-16 and 21-25, and still more
preferably a
TAT basic region peptide selected from the group set forth in SEQ ID NOS: 1-16
including one or more retroinverted analogs thereof e.g., as set forth in any
one of SEQ
ID NOS: 9-16. In a particularly preferred embodiment, the protein transduction
domain is a TAT basic region peptide comprising the sequence set forth in SEQ
ID
NO: 1 or a retroinverted TAT basic region peptide comprising the sequence set
forth in
SEQ ID NO: 9.

It will be apparent from the sequence data provided herein that the AP-1
signaling
inhibitory peptide may be separated from a protein transduction domain by one
or more
linkers, preferably a linker comprising from 1 to about 6 glycine residues or
other
amino acids of low immunogenicity e.g., serine. In a particularly preferred


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-21 -

embodiment, the protein transduction domain is separated from the AP-1
signaling
inhibitory peptide by at least one glycine residue which may be provided by
the
terminal amino acid of one or other peptide moiety.

In each of the foregoing embodiments by virtue of the small size of PhylomerTm
peptides, conservative amino acid variants of the exemplified peptides will
differ only
in 1 or 2 or 3 or 4 or 5 amino acid residues. Whilst such variants will
naturally possess
neuroprotective activity to retain their utility in performing an method of
the invention
described herein below, the present invention clearly encompasses any and all
such
1o variants in so far as they possess enhanced neuroprotective function in any
of the assay
systems exemplified herein. Preferred amino acid substitutions producing such
variants
are described herein.

The present invention clearly extends to any derivatives of the exemplified
neuroprotective peptides described in accordance with any one or more of the
foregoing examples.

In each of the embodiments described herein, it is preferred that all amino
acids in a
retroinverted peptide other than glycine are D-amino acids.


The present invention also provides a neuroprotective composition comprising
(i) an
amount of a neuroprotective AP-1 signaling inhibitory peptides according to
any one or
more embodiments described herein sufficient to reduce, delay or prevent
neuronal
apoptosis and/or necrosis in an animal; and (ii) a suitable carrier or
excipient for
application to the central nervous system of the animal.

Compositions comprising multiple neuroprotective AP-1 signaling inhibitory
peptides
are clearly contemplated herein, for enhanced benefit. It is preferred but not
essential
for such compositions to comprise active ingredients having different effects
or
3o activities in vivo. For example, suitable compositions may comprise
combinations of
combinations of peptides PYC19 and/or PYC35 and/or PYC36 and/or PYC 38/39


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-22-
and/or PYC41, and preferably a combination of PYC35 and PYC36, optionally in
further combination with the JNK inhibitory peptide JNK1-ID-TAT, and
preferably as
fusions with protein transduction domain(s) such as TAT basic region. Certain
linker
residues such as glycine may be found joining the protein transduction domain
with the
Phylomer peptides. Again, one or more of the constituent peptides may be
retroinverted.

As used herein, the term "amount sufficient to prevent or reduce neuronal
apoptosis
and/or necrosis in an animal" or similar term shall be taken to mean a
sufficient
1o quantity of a stated integer to reduce the number of neurons undergoing
apoptosis
and/or necrosis induced by a insult such as a physical injury e.g., acute
cortical injury,
or ischemic event in the animal. The precise amount of the stated integer will
vary
depending on the specific activity of the integer and/or the severity of the
insult.
Accordingly, this term is not to be construed to limit the invention to a
specific
quantity, e.g., weight or concentration, unless specifically stated otherwise.
Methods
for assessing efficacy of any amount of a peptide of the present invention in
preventing
neuronal cell death i.e., apoptosis and/or necrosis will be apparent to the
skilled artisan
from the disclosure herein.

2o As used herein, the term "suitable carrier or excipient" shall be taken to
mean a
compound or mixture thereof that is suitable for use in a formulation for
administration
to neurons or neuronal tissue albeit not necessarily limited in use to that
context.
Similarly, the term "carrier or excipient for neuronal application" shall be
taken to
mean a compound or mixture thereof that is suitable for application to
neuronal tissues
and which may be suitable for use in other contexts. In contrast, a "neuronal
tissue
carrier or excipient" is compound or mixture thereof that is described in the
art only
with reference to a use in formulations used on neuronal tissues.

A carrier or excipient useful in the composition of the present invention will
generally
3o not inhibit to any significant degree a relevant biological activity of the
active
compound e.g., the carrier or excipient will not significantly inhibit the AP-
1 signaling


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 23 -

inhibitory activity of the active peptide. For example, a carrier or excipient
may merely
provide a buffering activity to maintain the active compound at a suitable pH
to thereby
exert its biological activity, e.g., phosphate buffered saline. Alternatively,
or in
addition, the carrier or excipient may comprise a compound that enhances
neuronal
uptake of the peptide and/or enhances neuronal delivery. Alternatively, or in
addition,
the carrier or excipient may comprise a compound that enhances the activity or
half-life
of the active peptide e.g., a protease inhibitor. In yet another example, the
carrier or
excipient may include an antibiotic and/or an anaesthetic.

io Suitable carriers for use in protecting neurons in vivo include e.g., gels,
emulsions or
saline in which the peptide(s) is/are substantially soluble. Particularly
preferred
carriers are suitable for administration by injection to neuronal tissue or
alternatively,
by direct application to wounded neural tissue e.g., brain lesion.

In another example, the composition of the present invention comprises an
additional
composition of matter having synergistic activity with respect to the active
peptide in
so far as neuron repair is concerned e.g., an antioxidant compound and/or stem
cell.
The present invention also provides a method for producing a composition
described
2o herein according to any embodiment. For example, in its broadest form, such
a method
comprises mixing or otherwise combining an amount of an AP-1 signaling
inhibitory
peptide of the present invention sufficient to reduce or prevent neuronal cell
death in an
animal and a suitable carrier or excipient. In one example, the method
additionally
comprises producing or obtaining the AP-1 signaling inhibitory peptide. For
example,
a peptide inhibitor or a nucleic acid inhibitor is produced synthetically or
recombinantly, using a method known in the art and/or described herein.

The present invention also provides a method for preventing or delaying
neuronal cell
death in a subject comprising administering an AP-1 signaling inhibitory
peptide or
composition comprising said peptide according to any embodiment described
herein or
an analog of said peptide to a subject in need of treatment.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-24-
The present invention also provides a method for preventing or delaying
neuronal cell
death in a subject comprising administering a composition comprising one or
more
AP-1 signaling inhibitory peptides according to any embodiment described
herein to a
subject in need of treatment.

As used herein, the term "subject in need thereof' shall be taken to mean a
subject that
has developed or suffers from a neuronal condition involving e.g., glutamate
and/or
NMDA excitotoxicity and/or ischemia and/or wounding. For example, the subject
may
1o have recently suffered from, or is likely to suffer from, cerebral
ischemia, traumatic
brain injury, epilepsy, Parkinson's Disease, Alzheimer's Disease and
Amyotrophic
Lateral Sclerosis (ALS). In view of the efficacy of certain peptides early in
an
ischemic event e.g., within 1-7 days and preferably within 1-4 days or the day
following an ischemic event, the subject is more likely to be a subject having
recently
suffered local or global cerebral ischemia or traumatic brain injury e.g.,
following a
motor vehicle accident.

Similarly, neuronal cell death treatable by the present invention is
preferably induced
by NMDA excitotoxicity and/or glutamate excitotoxicity and/or ischemia and/or
wounding e.g., acute cortical injury.

In another example, an AP-1 signaling inhibitory peptide or composition is
administered by a process comprising administering nucleic acid encoding an AP-
1
signaling inhibitory peptide to a subject by particle bombardment under
conditions
sufficient for transcription and translation of said nucleic acid to occur.

The therapeutic method described herein is not to be limited to a single
application of a
peptide or composition of the invention. The present invention also
contemplates
repeated administration of a peptide or composition as described herein
according to
3o any embodiment e.g., to extend the period over which beneficial effects are
derived.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 25 -

In another example, the therapeutic method of the invention additionally
comprises
providing or obtaining a composition as described herein according to any
embodiment
or information concerning same. For example, the present invention provides a
method
of treatment of a subject in need thereof, said method comprising:
(i) identifying a subject suffering from or developing a neuronal condition
involving e.g., glutamate and/or NMDA excitotoxicity and/or ischemia and/or
wounding;
(ii) obtaining an AP-1 signaling inhibitory peptide or composition comprising
said
peptide as described herein according to any embodiment; and
lo (iii) administering said peptide or composition to said subject.

The present invention also provides a method of treatment of a subject in need
thereof,
said method comprising:
(i) identifying a subject suffering from or developing a neuronal condition
involving e.g., glutamate and/or NMDA excitotoxicity and/or ischemia and/or
wounding; and
(ii) recommending administration of AP-1 signaling inhibitory peptide or
composition comprising said peptide as described herein according to any
embodiment.
Alternatively, the method of treatment comprises administering or recommending
administration of AP-1 signaling inhibitory peptide or composition comprising
said
peptide as described herein according to any embodiment to a subject
previously
identified as suffering from a neuronal condition involving e.g., glutamate
and/or
NMDA excitotoxicity and/or ischemia and/or wounding.

The present invention also provides for the use of an amount of an AP-1
signaling
inhibitory peptide as described herein according to any embodiment sufficient
to inhibit
or delay neuronal cell death in the manufacture of a medicament for the
treatment of a
3o neuronal condition involving e.g., glutamate and/or NMDA excitotoxicity
and/or
ischemia and/or wounding.

Brief description of the drawings

Figure 1 is a graphical representation showing, on the y-axis the percentages
of viable
neurons in culture in the presence of 5 M extracellular concentration of the


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 26 -

Phylomerm peptides indicated on the x-axis (PYC 19L-TAT, PYC35L-TAT, PYC36L-
TAT, PYC38/39L-TAT and PYC41L-TAT) following incubation with glutamate to
induce excitotoxicity. Controls consisted of neuron cultures grown without
glutamate
(No insult), or without added peptide (Control), TAT-D peptide (TAT-D), JNK1-
1D-
TAT peptide (JNK1-ID-TAT), or neuron cultures incubated with a mixture of
glutamate receptor inhibitors (Blockers). MTS data were expressed as
percentage
neuronal viability with no insult control taken as 100% viability and insult
control as
5% viability (mean SEM; n=4; * p<0.005; **p<0.0001).

1o Figure 2 is a graphical representation showing, on the y-axis the
percentages of viable
neurons in culture in the presence of 5 M extracellular concentration of the
retroinverted PhylomerTM peptides indicated on the x-axis (PYC35L-TAT, PYC35L-
Scram-TAT, PYC36L-TAT and PYC36L Scram-TAT) following incubation with
glutamate to induce excitotoxicity. Controls consisted of neuron cultures
grown
without glutamate (No insult), or without added peptide (Control), or JNK1-ID-
TAT
peptide (JNK1-ID-TAT). Data show reduced neuroprotection when the PhylomerTm
sequences were scrambled. MTS data were expressed as percentage neuronal
viability
with no insult control taken as 100% viability and insult control as 5%
viability (mean
SEM; n=4; * p<0.005; * *p<0.0001).


Figure 3 shows copies of photomicrographs of neurons from cultures grown
without
glutamate (No insult), or incubated with glutamate in the presence of the
PhylomerTM
peptides (PYC19L-TAT, PYC35L-TAT, PYC36L-TAT, PYC38/39L-TAT and
PYC41L-TAT), a TAT-D peptide (TAT-D), the JNK1-ID-TAT peptide (JNKl-1D-
TAT), or without added peptide (Control), or with a mixture of glutamate
receptor
inhibitors (Blockers), as indicated in the top right of each panel.
Significantly higher
neuronal viability was observed for cultures not receiving glutamate, or
receiving
glutamate and incubated with the PhylomerTM peptides compared to TAT-D peptide
(TAT-D), or no added peptide. Magnification x 200.

Figure 4a provides graphical representations showing, on the y-axis the
percentages of


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-27-
viable neurons in culture in the presence of 0.1-10gM extracellular
concentration of the
PhylomerTm peptides PYC19L-TAT and PYC19D-TAT following incubation with
glutamate to induce excitotoxicity. Controls consisted of neuron cultures
grown
without glutamate (No insult), or without added peptide (Control). MTS data
were
expressed as percentage neuronal viability with no insult control taken as
100%
viability and insult control as 5% viability (mean f SEM; n=4; * p<0.005;
* *p<0.0001).

Figure 4b provides graphical representations showing, on the y-axis the
percentages of
i.o viable neurons in culture in the presence of 0.1-10 M extracellular
concentration of the
PhylomerTM peptides PYC35L-TAT and PYC35D-TAT following incubation with
glutamate to induce excitotoxicity. Controls consisted of neuron cultures
grown
without glutamate (No insult), or without added peptide (Control). MTS data
were
expressed as percentage neuronal viability with no insult control taken as
100%
viability and insult control as 5% viability (mean ~ SEM; n=4; * p<0.005;
* *p<0.0001).

Figure 4c provides graphical representations showing, on the y-axis the
percentages of
viable neurons in culture in the presence of 0.1-10 M extracellular
concentration of the
PhylomerTm peptides PYC36L-TAT and PYC36D-TAT following incubation with
glutamate to induce excitotoxicity. Controls consisted of neuron cultures
grown
without glutamate (No insult), or without added peptide (Control). MTS data
were
expressed as percentage neuronal viability with no insult control taken as
100%
viability and insult control as 5% viability (mean ~ SEM; n=4; * p<0.005;
* *p<0.0001).

Figure 4d provides graphical representations showing, on the y-axis the
percentages of
viable neurons in culture in the presence of 0.1-10gM extracellular
concentration of the
PhylomerTM peptides PYC38/39L-TAT and PYC38/39D-TAT following incubation
with glutamate to induce excitotoxicity. Controls consisted of neuron cultures
grown
without glutamate (No insult), or without added peptide (Control). MTS data
were


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 28-

expressed as percentage neuronal viability with no insult control taken as
100%
viability and insult control as 5% viability (mean ~ SEM; n=4; * p<0.005;
* *p<0.0001).

Figure 4e provides graphical representations showing, on the y-axis the
percentages of
viable neurons in culture in the presence of 0.1-10 M extracellular
concentration of the
PhylomerTM peptides PYC41L-TAT and PYC41D-TAT following incubation with
glutamate to induce excitotoxicity. Controls consisted of neuron cultures
grown
without glutamate (No insult), or without added peptide (Control). MTS data
were
io expressed as percentage neuronal viability with no insult control taken as
100%
viability and insult control as 5% viability (mean ~ SEM; n=4; * p<0.005;
* *p<0.0001).

Figure 5 provides a graphical representation showing, on the y-axis the
percentages of
viable neurons in culture in the presence of 5 M extracellular concentration
of the
Phylomer~Fm peptides PYC35D-TAT, PYC35L-FM, PYC35D-FM, PYC36D-TAT,
PYC36L-FM and PYC36D-FM, following incubation with glutamate to induce
excitotoxicity. Controls consisted of neuron cultures grown without glutamate
(No
insult), or without added peptide (Control). MTS data were expressed as
percentage
neuronal viability with no insult control taken as 100% viability and insult
control as
5% viability (mean SEM; n=4; * p<0.005; **p<0.0001).

Figure 6 provides a graphical representation showing, on the y-axis the
percentages of
viable neurons in culture in the presence of 5 M extracellular concentration
of the
PhylomerTM peptides PYC35D-TAT and PYC36D-TAT administered 15 min prior to
incubation with glutamate to induce excitotoxicity ("15 mins pre"), or
alternatively, at
the same time as incubation with glutamate ("0 mins") or 15 min following
incubation
with glutamate ("15 mins post"). Controls consisted of neuron cultures grown
without
glutamate (No insult), or without added peptide (Control), or neuron cultures
incubated
with glutamate in the presence of the peptide JNK1-ID-TAT or glutamate
receptor
inhibitors (Blockers) added 15 min prior to incubation with glutamate to
induce


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-29-
excitotoxicity ("15 mins pre"), at the same time as incubation with glutamate
("0
mins") or 15 min following incubation with glutamate ("15 mins post"). MTS
data
were expressed as percentage neuronal viability with no insult control taken
as 100%
viability and insult control as 5% viability (mean ~ SEM; n=4; * p<0.005;
**p<0.0001).

Figure 7a provides graphical representations showing the time course of
calcium influx
in cortical neurons exposed to glutamate (30 second time point; arrow) in the
presence
of PhylomerTM peptide PYC36D-TAT (lower panel), compared to neurons not
1o receiving glutamate (top panel) or receiving glutamate receptor inhibitors
(middle
panel).

Figure 7 b provides a graphical representation showing the amplitude (y-axis)
between
basal Ca2+ level and peak Ca2+ level for neuron cultures in the absence and
presence of
glutamate, for each condition indicated on the x-axis. Control: No added
peptide or
receptor inhibitor in the presence and absence of glutamate; Blockers:
glutamate
receptor inhibitors added in the presence and absence of glutamate; PYC5D-TAT,
the
PhylomerTM peptide PYC35D-TAT was added in the presence and absence of
glutamate; and PYC36D-TAT, the PhylomerTM peptide PYC36D-TAT was added in
the presence and absence of glutamate.

Figure 8 provides a graphical representation showing, on the y-axis the
percentages of
viable neurons in culture in the presence of 5gM extracellular concentration
of the
PhylomerTM peptides PYC35D-TAT and PYC36D-TAT administered 15 min prior to
incubation with NMDA to induce excitotoxicity. Controls consisted of neuron
cultures
grown without NMDA (No insult), or without added peptide (NMDA Control), or
neuron cultures incubated with NMDA in the presence of glutamate receptor
inhibitors
(Blockers). MTS data were expressed as percentage neuronal viability with no
insult
control taken as 100% viability and insult control as 5% viability (mean
SEM; n=4;
* p<0.005; **p<0.0001).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-30-
Figure 9 is a graphical representation showing the ability of retroinverted
PhylomerTm
peptides to protect cultured cortical neurons in an in vitro model of ischemia
known as
Oxygen Glucose Deprivation (OGD). Neuronal survival (5) (y-axis) was
determined in
the absence ("No IVI control") or following 35 min OGD in the absence of
exogenously-added peptide ("IVI Control") or in the presence of peptides
PYC35D-
TAT or PYC36D-TAT added before ("pre") or after ("post") commencement of OGD.
Figure 10 is a graphical representat"ion showing the stability of
retroinverted
Phylomer~ peptides in serum. Peptide concentration ( M) is shown on the y-axis
at
xo various time points (min) after administration to animals as indicated on
the x-axis.
Neuronal survival (y-axis) was determined in the absence ("No IVI control") or
following 35 min OGD in the absence of exogenously-added peptide ("IVI
Control") or
in the presence of peptides PYC35D-TAT or PYC36D-TAT added before ("pre") or
after ("post") commencement of OGD.

Figure 11 is a graphical representation showing the extent of neuronal loss in
the
perilesion area I day post-injury following administration of Phylomerm
peptides
PYC35D-TAT and PYC36D-TAT, and the peptide JNKl-1D-TAT, as determined by
hippocampal CAl cell count. As a negative control, hippocampal CAl cell count
was
2o determined following administration of PYC35D Scram-TAT peptide, containing
the
scrambled sequence of PYC35D was employed. Hippocampal CA1 cell counts were
also determined for sham-treated animals, and for animals receiving saline
(NaCI).
Values are expressed as the loss of MAP2 immunoreactive area (mm2).

Figure 12 is a graphical representation showing the extent of neuronal loss in
the
perilesion area 1 day post-injury following administration of PhylomerTm
peptides
PYC35D-TAT and PYC36D-TAT, and the peptide JNK1-ID-TAT, as determined by
loss of MAP immunoreactivity (mm) 1 day post injury following administration
of
PhylomerTM peptide PYC35D-TAT, the negative control peptide PYC35D Scram-TAT,
or the peptide JNKl-1D-TAT.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-31-
Figure 13 is a copy of a photomicrograph showing loss of MAP immunoreactivity
(area
enclosed by dotted line) in perilesion area 1 day post injury following
administration of
PhylomerTm PYC35D-TAT or negative control peptide (PYC35D Scram-TAT). Size
bar 25 m.

Detailed description of the preferred embodiments
Peptide inhibitors ofAP-1 signaling
As exemplified herein, the present inventors have identified a number of
1o neuroprotective AP-1 signaling inhibitory peptides that partially or
completely inhibit
c-Jun homodimerization and/or c-Jun heterodimerization and/or one or more
upstream
signaling steps in the AP-1 signaling pathway e.g., Cdc42 and/or Racl and/or
Pakl
and/or MKK (Table 1), thereby preventing, delaying or reducing neuronal cell
death by
apoptotic and/or necrotic pathways. The peptides partially or completely
decrease,
prevent or inhibit neuronal cell death mediated by glutamate excitotoxicity
and/or
NMDA excitotoxicity, albeit not necessarily at the level of the glutamate/NMDA
receptor. Additionally, the peptides have been shown herein to be
neuroprotective in
models of local and global ischemia in vitro as well as in vivo.

2o The present invention clearly extends to variants of the exemplified
neuroprotective
AP-1 signaling inhibitory peptides, such as derivatives and/or analogs, by
modification
to the sequences provided herein. The invention also extends to homologs i.e.,
functionally-equivalent peptides having related sequences to the sequences
provided
herein e.g., using different expression libraries to those used as a source of
the
neuroprotective peptides described herein.

It is understood by the skilled artisan that, inherent in the definition of a
biologically
functional equivalent protein or peptide, is the concept that there is a limit
to the
number of changes that may be made within a defined portion of the molecule
and still
result in a molecule with an acceptable level of equivalent biological
activity.
Biologically functional equivalent peptides are thus defined herein as those
peptides in


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-32-
which specific amino acids may be substituted or deleted. Particular
embodiments
encompass variants that have one, two, three, four, five or more variations in
the amino
acid sequence relative to a base peptide subject to the retention of
neuroprotective
function in one or more assays described in the examples. Of course, a
plurality of
variants may be made and used in accordance with the invention.
Peptide derivatives
As used herein the term "derivative" shall be taken to mean a peptide that is
derived
from an AP-1 signaling inhibitory peptide exemplified herein e.g., a fragment
or
1o processed form of the peptide, or a molecule comprising one or more amino
acid
substitutions, or comprising additional amino acid residues or non-amino acid
substituents, relative to the base peptide from which it is derived. The term
"derivative" also encompasses fusion proteins comprising a peptide of the
invention.

Exemplary fusion protein comprises a label, such as, for example, an epitope,
e.g., a
FLAG epitope or a V5 epitope or an HA epitope. Such a tag is useful for, for
example,
purifying the fusion protein.

A "conservative amino acid substitution" is one in which an amino acid residue
is
2o replaced with another amino acid residue without disturbing the overall
structure of the
peptide. Such changes tend to rely on similarity in hydrophilicity and/or
polarity of the
substituent. The size and/or charge of the side chains also are relevant
factors in
determining which substitutions are conservative. Families of amino acid
residues
having similar side chains have been defined in the art, including basic side
chains
(e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid,
glutamic acid),
uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine,
threonine,
tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine,
isoleucine,
proline, phenylalanine, methionine, tryptophan), .beta.-branched side chains
(e.g.,
threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine,
phenylalanine,
tryptophan, histidine).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-33-
Those skilled in the art are well aware that the following substitutions are
permissible
conservative substitutions (i) substitutions involving arginine, lysine and
histidine; (ii)
substitutions involving alanine, glycine and serine; and (iii) substitutions
involving
phenylalanine, tryptophan and tyrosine.

The importance of the hydropathic amino acid index in conferring interactive
biological
function on a protein is generally understood in the art (Kyte & Doolittle, J.
Mol. Biol.
157, 105-132, 1982). It is known that certain amino acids may be substituted
for other
amino acids having a similar hydropathic index or score and still retain a
similar
io biological activity. The hydropathic index of amino acids also may be
considered in
determining a conservative substitution that produces a functionally
equivalent
molecule. Each amino acid has been assigned a hydropathic index on the basis
of their
hydrophobicity and charge characteristics, as follows: isoleucine (+4.5);
valine (+4.2);
leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine
(+1.9);
alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-
0.9); tyrosine
(-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5);
aspartate (-
3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5). In making changes
based
upon the hydropathic index, the substitution of amino acids whose hydropathic
indices
are within .+/- 0.2 is preferred. More preferably, the substitution will
involve amino
2o acids having hydropathic indices within .+/- 0.1, and more preferably
within about +/-
0.05.

It is also understood in the art that the substitution of like amino acids is
made
effectively on the basis of hydrophilicity, particularly where the biological
functional
equivalent protein or peptide thereby created is intended for use in
immunological
embodiments, as in the present case (e.g. US Patent No. 4,554,101), In fact,
the greatest
local average hydrophilicity of a protein, as governed by the hydrophilicity
of its
adjacent amino acids, correlates with its immunogenicity and antigenicity. As
detailed
in US Patent No. 4,554,101, the following hydrophilicity values have been
assigned to
3o amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 +/-
0.1); glutamate
(+3.0 +/- 0.1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine
(0); threonine


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-34-
(-0.4); proline (-0.5 +/- 0.1); alanine (-0.5); histidine (-0.5); cysteine (-
1.0); methionine
(-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3);
phenylalanine (-
2.5); tryptophan (-3.4). In making changes based upon similar hydrophilicity
values, it
is preferred to substitute amino acids having hydrophilicity values within
about +/- 0.2
of each other, more preferably within about +/- 0.1, and even more preferably
within
about +/- 0.05

Non-amino acid substituents may be linked covalently to a peptide e.g., via an
amino
terminal amino acid residue, a carboxy terminal amino acid residue, or at an
internal
1o amino acid residue. Such modifications include the addition of a protective
or capping
group on a reactive moiety in the peptide, addition of a detectable label, and
other
changes that do not adversely destroy the activity of the peptide compound.
For
example, particular peptide residues may be derivatized or chemically modified
in
order to enhance the stability of the peptide or to permit coupling of the
peptide to other
agents, particularly lipids.

Chemical moieties may be linked covalently to a peptidyl moiety e.g., via an
amino
terminal amino acid residue, a carboxy terminal amino acid residue, or at an
internal
amino acid residue. Such modifications include the addition of a protective or
capping
group on a reactive moiety in the peptide, addition of a detectable label, and
other
changes that do not adversely destroy the activity of the peptide compound.

An "amino terminal capping group" of a peptide described herein is any
chemical
compound or moiety that is covalently linked or conjugated to the amino
terminal
amino acid residue of a peptide compound. An amino terminal capping group may
be
useful to inhibit or prevent intramolecular cyclization or intermolecular
polymerization,
to promote transport of the peptide compound across the blood-brain barrier
(BBB), to
protect the amino terminus from an undesirable reaction with other molecules,
to
provide additional antioxidative activity, or to provide a combination of
these
properties. A peptide compound of this invention that possesses an amino
terminal
capping group may possess other beneficial activities as compared with the
uncapped


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 35 -

peptide, such as enhanced efficacy or reduced side effects. Examples of amino
terminal
capping groups that are useful in preparing peptide compounds and compositions
according to this invention include, but are not limited to, 1 to 6 naturally
occurring L-
amino acid residues, preferably, 1-6 lysine residues, = 1-6 arginine residues,
or a
combination of lysine and arginine residues; urethanes; urea compounds; lipoic
acid
("Lip"); glucose-3-O-glycolic acid moiety ("Gga"); or an acyl group that is
covalently
linked to the amino terminal amino acid residue of a peptide, wherein such
acyl groups
useful in the compositions of the invention may have a carbonyl group and a
hydrocarbon chain that ranges from one carbon atom (e.g., as in an acetyl
moiety) to up
io to 25 carbons (e.g., palmitoyl group, "Palm" (16:0) and docosahexaenoyl
group,
"DHA" (C22:6-3)). Furthermore, the carbon chain of the acyl group may be
saturated,
as in Palm, or unsaturated, as in DHA. It is understood that when an acid,
such as
docosahexaenoic acid, palmitic acid, or lipoic acid is designated as an amino
terminal
capping group, the resultant peptide compound is the condensed product of the
uncapped peptide and the acid.

A "carboxy terminal capping group" of a peptide compound described herein is
any
chemical compound or moiety that is covalently linked or conjugated to the
carboxy
terminal amino acid residue of the peptide compound. The primary purpose of
such a
carboxy terminal capping group is to inhibit or prevent intramolecular
cyclization or
intermolecular polymerization, to promote transport of the peptide compound
across
the blood-brain barrier, and to provide a combination of these properties. A
peptide
compound of this invention possessing a carboxy terminal capping group may
also
possess other beneficial activities as compared with the uncapped peptide,
such as
enhanced efficacy, reduced side effects, enhanced hydrophilicity, enhanced
hydrophobicity. Carboxy terminal capping groups that are particularly useful
in the
peptide compounds described herein include primary or secondary amines that
are
linked by an amide bond to the .alpha.-carboxyl group of the carboxy terminal
amino
acid of the peptide compound. Other carboxy terminal capping groups useful in
the
invention include aliphatic primary and secondary alcohols and aromatic
phenolic
derivatives, including flavenoids, with 1 to 26 carbon atoms, which form
esters when


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-36-
linked to the carboxylic acid group of the carboxy terminal amino acid residue
of a
peptide compound described herein.

Other chemical modifications of a peptide or analog, include, for example,
glycosylation, acetylation (including N-terminal acetylation), carboxylation,
carbonylation, phosphorylation, PEGylation, amidation, addition of trans
olefin,
substitution of a-hydrogens with methyl groups, derivatization by known
protecting/blocking groups, circularization, inhibition of proteolytic
cleavage (e.g.,
using D amino acids), linkage to an antibody molecule or other cellular
ligand, etc. Any
io of numerous chemical modifications may be carried out by known techniques,
including but not limited to specific chemical cleavage by cyanogen bromide,
trypsin,
chymotrypsin, papain, V8 protease, NaBH4, acetylation, formylation, oxidation,
reduction, etc.

Peptide analogs
In another example of the invention, an AP-1 signaling inhibitory peptide
analog is
prepared. As used herein, the term "analog" shall be taken to mean a peptide
that is
modified to comprise one or more non-naturally-occurring amino acids.

2o Analogs may also comprise sterically similar compounds that mimic critical
subdomains of a peptide. Such "peptidomimetics" are produced by modelling and
chemical design processes known to those of skill in the art.

Preferred analogs of an AP-1 signaling inhibitory peptides comprise one or
more non-
naturally occurring amino acids or amino acid analogs. For example, a peptide
inhibitor as described herein comprises one or more naturally occurring non-
genetically
encoded L-amino acids, synthetic L-amino acids or D-enantiomers of an amino
acid.
For example, the peptide comprises only D-amino acids. For example, the analog
comprises one or more residues selected from the group consisting of:
hydroxyproline,
0-alanine, 2,3-diaminopropionic acid, a-aminoisobutyric acid, N-methylglycine
(sarcosine), omithine, citrulline, t-butylalanine, t-butylglycine, N-
methylisoleucine,


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-37-
phenylglycine, cyclohexylalanine, norleucine, naphthylalanine, pyridylananine
3-
benzothienyl alanine 4-chlorophenylalanine, 2-fluorophenylalanine, 3-
fluorophenylalanine, 4-fluorophenylalanine, penicillamine, 1,2,3,4-tetrahydro-
tic
isoquinoline-3-carboxylic acid 0-2-thienylalanine, methionine sulfoxide,
homoarginine,
N-acetyl lysine, 2,4-diamino butyric acid, p-aminophenylalanine , N-
methylvaline,
homocysteine, homoserine, E-amino hexanoic acid, 8-amino valeric acid, 2,3-
diaminobutyric acid and mixtures thereof.

Other amino acid residues that are useful for making the peptides and peptide
analogs
1o described herein can be found, e.g., in Fasman, 1989, CRC Practical
Handbook of
Biochemistry and Molecular Biology, CRC Press, Inc., and the references cited
therein.
The present invention additionally encompasses an isostere of a peptide
described
herein. The term "isostere" as used herein is intended to include a chemical
structure
that can be substituted for a second chemical structure because the steric
conformation
of the first structure fits a binding site specific for the second structure.
The term
specifically includes peptide back-bone modifications (i.e., amide bond
mimetics)
known to those skilled in the art. Such modifications include modifications of
the
amide nitrogen, the a-carbon, amide carbonyl, complete replacement of the
amide
2o bond, extensions, deletions or backbone crosslinks. Several peptide
backbone
modifications are known, including yr[CH2S], W[CH2NH], yV[CSNH2], yi[NHCO],
yr[COCHz], and yr[(E) or (Z) CH=CH]. In the nomenclature used above, yf
indicates the
absence of an amide bond. The structure that replaces the amide group is
specified
within the brackets.
Other modifications include, for example, an N-alkyl (or aryl) substitution
(yr
[CONR]), or backbone crosslinking to construct lactams and other cyclic
structures.
Other derivatives of the nzodulator compounds of the invention include C-
terminal
hydroxymethyl derivatives, 0-modified derivatives (e.g., C-terminal
hydroxymethyl
3o benzyl ether), N-terminally modified derivatives including substituted
amides such as
alkylamides and hydrazides.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 38 -

In another example, a peptide analog is a retro peptide (see, for example,
Goodman et
al., Accounts of Chemical Research, 12:1-7, 1979). A retro peptide comprises a
reversed amino acid sequence of a peptide inhibitor described herein.
Optionally, the
retro peptide analog comprises an additional feature, such as, for example, a
protein
transduction domain, which may also be a retro peptide.

In a further example, an analog of a peptide described herein is a retro-
inverso peptide
(Sela and Zisman, FASEB J. 11:449, 1997). Evolution has ensured the almost
exclusive
1o occurrence of L-amino acids in naturally occurring proteins. As a
consequence,
virtually all proteases cleave peptide bonds between adjacent L- amino acids.
Accordingly, artificial proteins or peptides composed of D-amino acids are
preferably
resistant to proteolytic breakdown. Retro-inverso peptide analogs are isomers
of linear
peptides in which the direction of the amino acid sequence is reversed (retro)
and the
chirality, D- or L-, of one or more amino acids therein is inverted (inverso)
e.g., using
D-amino acids rather than L-amino acids, e.g., Jameson et al., Nature, 368,
744-746
(1994); Brady et al., Nature, 368, 692-693 (1994). The net result of combining
D-
enantiomers and reverse synthesis is that the positions of carbonyl and amino
groups in
each amide bond are exchanged, while the position of the side-chain groups at
each
2o alpha carbon is preserved. An advantage of retro-inverso peptides is their
enhanced
activity in vivo due to improved resistance to proteolytic degradation, i.e.,
the peptide
has enhanced stability. (e.g., Chorev et al., Trends Biotech. 13, 438-445,
1995).
Retro-inverso or retroinverted peptide analogs may be complete or partial.
Complete
retro-inverso peptides are those in which a complete sequence of a peptide
described
herein is reversed and the chirality of each amino acid other than glycine in
a sequence
is inverted. The exclusion of glycine is based on the fact that glycine does
not have a
chiral analog. Partial retro-inverso peptide analogs are those in which only
some of the
peptide bonds are reversed and the chirality of only those amino acid residues
in the
3o reversed portion is inverted. For example, one or two or three or four or
five or six or
seven or eight or nine or ten or eleven or twelve or thirteen or fourteen or
fifteen or


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-39-
sixteen or seventeen or eighteen or nineteen or twenty or twenty one or twenty
two or
twenty three or twenty four or twenty five or twenty six or twenty seven or
twenty eight
or twenty nine or thirty or thirty one or thirty two or thirty three or thirty
four or thirty
five or thirty six or thirty seven or thirty eight amino acid residues are D-
amino acids.
The present invention clearly encompasses both partial and complete retro-
inverso
peptide analogs. Such a retroinverso peptide analog may optionally include an
additional component, such as, for example, a protein transduction domain,
which may
also be retroinverted.

1o In one embodiment, the retro-inverso peptide is N-terminally modified, for
example,
with a modifying group comprising an alkyl group such as a Cl-C6 lower alkyl
group,
e.g., a methyl, ethyl, or propyl group; or a cyclic, heterocyclic, polycyclic
or branched
alkyl group, or one or more an amino acid linker residues.

In another embodiment, the retro-inverso peptide is C-terminally modified, for
example
with an amide group, an alkyl or aryl amide group (e.g., phenethylamide) or a
hydroxy
group (i.e., the reduction product of a peptide acid, resulting in a peptide
alcohol), or
one or more an amino acid linker residues e.g., glycine, cysteine, etc.

It is also within the scope of the present invention for the retro-inverso
peptide to be
further modified by the inclusion of one or more targeting domains e.g.,
penetratin,
TAT etc added to the N-terminus and/or C-terminus. Such peptide additions may
be
separated from the retro-inverso peptide moiety by one or more linkers e.g.,
glycine,
serine, cysteine, etc.

Protein transduction domains
To facilitate peptide entry into a cell, the peptide may be conjugated to
(e.g., expressed
as a fusion with) a protein transduction domain. As used herein, the term
"protein
transduction domain" shall be taken to mean a peptide or protein that is
capable of
3o enhancing, increasing or assisting penetration or uptake of a compound
conjugated to
the protein transduction domain into a cell either in vitro or in vivo. Those
skilled in


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-40-
the art will be aware that synthetic or recombinant peptides can be delivered
into cells
through association with a protein transduction domain such as the TAT
sequence from
HIV or the Penetratin sequence from the Antennapaedia homeodomain protein
(see, for
example, Temsamani and Vidal, Drug Discovery Today 9: 1012-1019, 2004, for
review).

A suitable protein transduction domain will be apparent to the skilled artisan
and
includes, for example, native conformations and retroinverted forms of HN-1
TAT
basic region (e.g., SEQ ID NOS: 1-16), Kaposi fibroblast growth factor (FGF)
protein
1o transduction domain (e.g., SEQ ID NOS: 17-20), signal sequence based
peptide 1(e.g.,
SEQ ID NO: 21), signal sequence based peptide 2 (e.g., SEQ ID NO: 22),
transportan
(e.g., SEQ ID NO: 23), amphiphilic model peptide (e.g., SEQ ID NO: 24) or
polyarginine (e.g., SEQ ID NO: 25).

Additional suitable protein transduction domains are described, for example,
by Zhao
and Weisledder Medicinal Research Reviews, 24: 1-12, 2004; or by Wagstaff and
Jans,
Current Medicinal Chemistry, 13: 1371-1387, 2006.

Linkers
2o The "core" AP-1 signaling inhibitory Phylomer~ peptide (e.g., identified
e.g., by
virtue of its ability to block c-Jun dimerization in yeast and/or to block AP-
1 regulated
luciferase reporter gene expression in mammalian cells) may be linked to
another
peptidyl moiety (e.g., for immunodetection such as a FLAG epitope, or for
targeting
such as a protein transduction domain), albeit separated there from by a
linker.
Preferred linkers facilitate the independent folding of each peptidyl moiety
in the
assembled AP-1 signaling inhibitory peptide, thereby reducing steric hindrance
of one
moiety by another moiety. The amino acid composition of a linker peptide is
important
for stability and folding of a fusion protein, rather than a specific sequence
(Robinson
3o and Sauer Proc. Natl. Acad. Sci. 95: 5929-5934, 1998).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-41-
Suitable linkers will be apparent to the skilled artisan and are predominantly
hydrophilic, i.e. the residues in the linker are hydrophilic.

It is also often unfavourable to utilize a linker sequence having a high
propensity to
adopt a-helix or P-strand structures, which could limit the flexibility of the
peptidyl
moieties and reduce functionality. Accordingly, preferred linkers may have a
preference to adopt extended conformations.

Preferred linkers comprise a high content of glycine and/or serine residues.
Linkers
1o comprising glycine and/or serine have a high freedom degree for linking of
two
proteins, i.e., they enable the fused proteins to fold and produce functional
proteins.
Glycine-rich linkers are particularly preferred because they force the linker
to adopt a
loop conformation. The absence of aP-carbon from glycine also permits the
polypeptide backbone to access dihedral angles that are energetically
forbidden for
other amino acids. A particularly preferred linker in the present context
consists of
polyglycine i.e., between about 2 and 6 glycine residues, or a single glycine
residue.
Chemical synthesis ofpeptides and peptide analogs
2o AP-1 signaling inhibitory peptides and any derivatives, analogs or homologs
thereof
are readily synthesized from their determined amino acid sequences using
standard
techniques, e.g., using BOC or FMOC chemistry. Synthetic peptides are prepared
using known techniques of solid phase, liquid phase, or peptide condensation,
or any
combination thereof, and can include natural and/or unnatural amino acids.
Amino
acids used for peptide synthesis may be standard Boc (Na-amino protected Na-t-
butyloxycarbonyl) amino acid resin with the deprotecting, neutralization,
coupling and
wash protocols of the original solid phase procedure of Merrifield, J. Am.
Chem. Soc.,
85:2149-2154, 1963, or the base-labile Na-amino protected 9-
fluorenylrnethoxycarbonyl (Fmoc) amino acids described by Carpino and Han, .I.
Org.
Chem., 37:3403-3409, 1972. Both Fmoc and Boc Na-amino protected amino acids
can
be obtained from various commercial sources, such as, for example, Fluka,
Bachem,


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-42-
Advanced Chemtech, Sigma, Cambridge Research Biochemical, Bachem, or Peninsula
Labs.

The Merrifield method of synthesis (Merrifield, JAm Chem Soc, 85,:2149-2154,
1963)
and the myriad of available improvements on that technology are described in
the art
(see e.g., Synthetic Peptides: A User's Guide, Grant, ed. (1992) W.H. Freeman
& Co.,
New York, pp. 382; Jones (1994) The Chemical Synthesis of Peptides, Clarendon
Press, Oxford, pp. 230.); Barany, G. and Merrifield, R.B. (1979) in The
Peptides
(Gross, E. and Meienhofer, J. eds.), vol. 2, pp. 1-284, Academic Press, New
York;
1o Wiinsch, E., ed. (1974) Synthese von Peptiden in Houben-Weyls Metoden der
Organischen Chemie (Muler, E., ed.), vol. 15, 4th edn., Parts 1 and 2, Thieme,
Stuttgart; Bodanszky, M. (1984) Principles of Peptide Synthesis, Springer-
Verlag,
Heidelberg; Bodanszky, M. & Bodanszky, A. (1984) The Practice of Peptide
Synthesis,
SpringeN-Ijerlag, Heidelberg; Bodanszky, M. (1985) Int. J. Peptide Protein
Res. 25,
449-474.

Generally, chemical synthesis methods comprise the sequential addition of one
or more
amino acids to a growing peptide chain. Normally, either the amino or carboxyl
group
of the first amino acid is protected by a suitable protecting group. The
protected or
2o derivatized amino acid can then be either attached to an inert solid
support or utilized in
solution by adding the next amino acid in the sequence having the
complementary
(amino or carboxyl) group suitably protected, under conditions that allow for
the
formation of an amide linkage. The protecting group is then removed from the
newly
added amino acid residue and the next amino acid (suitably protected) is then
added,
and so forth. After the desired amino acids have been linked in the proper
sequence,
any remaining protecting groups (and any solid support, if solid phase
synthesis
techniques are used) are removed sequentially or concurrently, to render the
final
polypeptide. By simple modification of this general procedure, it is possible
to add
more than one amino acid at a time to a growing chain, for example, by
coupling
(under conditions which do not racemize chiral centers) a protected tripeptide
with a
properly protected dipeptide to form, after deprotection, a pentapeptide. See,
e.g., J. M.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-43-
Stewart and J. D. Young, Solid Phase Peptide Synthesis (Pierce Chemical Co.,
Rockford, IL 1984) and G. Barany and R. B.Merrifield, The Peptides : Analysis,
Synthesis, Biology, editors E. Gross and J. Meienhofer, Vol. 2, (Academic
Press, New
York, 1980), pp. 3-254, for solid phase peptide synthesis techniques; and M.
Bodansky,
Principles of Peptide Synthesis, (Springer-Verlag, Berlin 1984)and E. Gross
and J.
Meienhofer, Eds. , The Peptides : Analysis. Synthesis. Biology, Vol.1, for
classical
solution synthesis. These methods are suitable for synthesis of a peptide of
the present
invention or an analog or derivative thereof.

1o Typical protecting groups include t-butyloxycarbonyl (Boc), 9-
fluorenylmethoxycarbonyl (Fmoc) benzyloxycarbonyl (Cbz); p-toluenesulfonyl
(Tx);
2,4-dinitrophenyl ; benzyl (Bzl); biphenylisopropyloxycarboxy-carbonyl, t-
amyloxycarbonyl, isobornyloxycarbonyl, o-bromobenzyloxycarbonyl, cyclohexyl,
isopropyl, acetyl, o-nitrophenylsulfonyl and the like.

Typical solid supports are cross-linked polymeric supports. These can include
divinylbenzene cross-linked-styrene-based polymers, for example,
divinylbenzene-
hydroxymethylstyrene copolymers, divinylbenzene- chloromethylstyrene
copolymers
and divinylbenzene-benzhydrylaminopolystyrene copolymers.


A peptide, analog or derivative as described herein can also be chemically
prepared by
other methods such as by the method of simultaneous multiple peptide
synthesis. See,
e. g. , Houghten Proc. Natl. Acad. Sci. USA 82: 5131-5135, 1985 or U. S.
Patent No.
4,631, 211.

Synthetic peptides may also be produced using techniques known in the art and
described, for example, in Stewart and Young (In: Solid Phase Synthesis,
Second
Edition, Pierce Chemical Co., Rockford, Ill. (1984) and/or Fields and Noble
(Int. J.
Pept. Protein Res., 35:161-214, 1990), or using automated synthesizers.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-44-
PhylomerTm peptides may also be produced in synthetic form as true cyclic
pepides.
Alternatively they may be cyclized following synthesis via the formation of a
covalent
bond between the termini such as the oxidation of flanking cystein residues or
by the
formation of a thioester or peptidyl bond.

Recombinant peptide production

AP-1 signaling inhibitory peptides of the present invention and any
derivatives, analogs
or homologs thereof are readily synthesized by recombinant means using methods
known in the art. For example, nucleic acid encoding a peptide is synthesized
from the
io deduced amino acid sequence (e.g., as set forth in Table 1).

To facilitate the production of a recombinant peptide or fusion protein
nucleic acid
encoding same is preferably isolated or synthesized. Typically the nucleic
acid
encoding the constituent components of the fusion protein is/are isolated
using a known
method, such as, for example, amplification (e.g., using PCR or splice overlap
extension) or isolated from nucleic acid from an organism using one or more
restriction
enzymes or isolated from a library of nucleic acids. Methods for such
isolation will be
apparent to the ordinary skilled artisan and/or described in Ausubel et al
(In: Current
Protocols in Molecular Biology. Wiley Interscience, ISBN 047 150338, 1987),
Sambrook et al (In: Molecular Cloning: Molecular Cloning: A Laboratory Manual,
Cold Spring Harbor Laboratories, New York, Third Edition 2001).

For expressing protein by recombinant means, a protein-encoding nucleotide
sequence
is placed in operable connection with a promoter or other regulatory sequence
capable
of regulating expression in a cell-free system or cellular system. For
example, nucleic
acid comprising a sequence that encodes a peptide in operable connection with
a
suitable promoter is expressed in a suitable cell for a time and under
conditions
sufficient for expression to occur. Nucleic acid encoding a peptide is readily
derived
from the publicly available amino acid sequence.



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 45 -

As used herein, the term "promoter" is to be taken in its broadest context and
includes
the transcriptional regulatory sequences of a genomic gene, including the TATA
box or
initiator element, which is required for accurate transcription initiation,
with or without
additional regulatory elements (e.g., upstream activating sequences,
transcription factor
binding sites, enhancers and silencers) that alter expression of a nucleic
acid (e.g., a
transgene), e.g., in response to a developmental and/or external stimulus, or
in a tissue
specific manner. In the present context, the term "promoter" is also used to
describe a
recombinant, synthetic or fusion nucleic acid, or derivative which confers,
activates or
enhances the expression of a nucleic acid (e.g., a transgene and/or a
selectable marker
io gene and/or a detectable marker gene) to which it is operably linked.
Preferred
promoters can contain additional copies of one or more specific regulatory
elements to
further enhance expression and/or alter the spatial expression and/or temporal
expression of said nucleic acid.

As used herein, the term "in operable connection with" "in connection with" or
"operably linked to" means positioning a promoter relative to a nucleic acid
(e.g., a
transgene) such that expression of the nucleic acid is controlled by the
promoter. For
example, a promoter is generally positioned 5' (upstream) to the nucleic acid,
the
expression of which it controls. To construct heterologous promoter/nucleic
acid
combinations (e.g., promoter/transgene and/or promoter/selectable marker gene
combinations), it is generally preferred to position the promoter at a
distance from the
gene transcription start site that is approximately the same as the distance
between that
promoter and the nucleic acid it controls in its natural setting, i.e., the
gene from which
the promoter is derived. As is known in the art, some variation in this
distance can be
accommodated without loss of promoter function.

Should it be preferred that a peptide or fusion protein of the invention is
expressed in
vitro a suitable promoter includes, but is not limited to a T3 or a T7
bacteriophage
promoter (Hanes and Pluckthun Proc. Natl. Acad. Sci. USA, 94 4937-4942 1997).



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-46-
Typical expression vectors for in vitro expression or cell-free expression
have been
described and include, but are not limited to the TNT T7 and TNT T3 systems
(Promega), the pEXP1-DEST and pEXP2-DEST vectors (Invitrogen).

Typical promoters suitable for expression in bacterial cells include, but are
not limited
to, the lacz promoter, the Ipp promoter, temperature-sensitive kL or kR
promoters, T7
promoter, T3 promoter, SP6 promoter or semi-artificial promoters such as the
IPTG-
inducible tac promoter or lacUV5 promoter. A number of other gene construct
systems
for expressing the nucleic acid fragment of the invention in bacterial cells
are well-
io known in the art and are described for example, in Ausubel et al (In:
Current Protocols
in Molecular Biology. Wiley Interscience, ISBN 047 150338, 1987), US Patent
No.
5,763,239 (Diversa Corporation) and Sambrook et al (In: Molecular Cloning:
Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratories, New
York, Third Edition 2001).

Numerous expression vectors for expression of recombinant polypeptides in
bacterial
cells and efficient ribosome binding sites have been described, and include,
for
example, PKC30 (Shimatake and Rosenberg, Nature 292, 128, 1981); pKKl73-3
(Amann and Brosius, Gene 40, 183, 1985), pET-3 (Studier and Moffat, J. Mol.
Biol.
2o 189, 113, 1986); the pCR vector suite (Invitrogen), pGEM-T Easy vectors
(Promega),
the pL expression vector suite (Invitrogen) the pBAD/TOPO or pBAD/thio - TOPO
series of vectors containing an arabinose-inducible promoter (Invitrogen,
Carlsbad,
CA), the latter of which is designed to also produce fusion proteins with a
Trx loop for
conformational constraint of the expressed protein; the pFLEX series of
expression
vectors (Pfizer nc., CT,USA); the pQE series of expression vectors (QIAGEN,
CA,
USA), or the pL series of expression vectors (Invitrogen), amongst others.

Typical promoters suitable for expression in viruses of eukaryotic cells and
eukaryotic
cells include the SV40 late promoter, SV40 early promoter and cytomegalovirus
(CMV) promoter, CMV IE (cytomegalovirus immediate early) promoter amongst
others. Preferred vectors for expression in mammalian cells (e.g., 293, COS,
CHO,


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-47-
lOT cells, 293T cells) include, but are not limited to, the pcDNA vector suite
supplied
by Invitrogen, in particular pcDNA 3.1 myc-His-tag comprising the CMV promoter
and encoding a C-terminal 6xHis and MYC tag; and the retrovirus vector
pSRatkneo
(Muller et al., Mol. Cell. Biol., 11, 1785, 1991).

A wide range of additional host/vector systems suitable for expressing a
peptide or
fusion protein of the present invention are available publicly, and described,
for
example, in Sambrook et al (In: Molecular cloning, A laboratory manual, second
edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989).
Means for introducing the isolated nucleic acid molecule or a gene construct
comprising same into a cell for expression are well-known to those skilled in
the art.
The technique used for a given organism depends on the known successful
techniques.
Means for introducing recombinant DNA into cells include microinjection,
transfection
mediated by DEAE-dextran, transfection mediated by liposomes such as by using
lipofectamine (Gibco, MD, USA) and/or cellfectin (Gibco, MD, USA), PEG-
mediated
DNA uptake, electroporation and microparticle bombardment such as by using DNA-

coated tungsten or gold particles (Agracetus Inc., WI, USA) amongst others.

Peptide/analog isolation
After being produced or synthesized, a peptide compound that is useful in the
compositions and methods of the invention may be purified using methods known
in
the art. Such purification preferably provides a peptide of the invention in a
state
dissociated from significant or detectable amounts of undesired side reaction
products;
unattached or unreacted moieties used to modify the peptide compound; and
dissociated from other undesirable molecules, including but not limited to
other
peptides, proteins, nucleic acids, lipids, carbohydrates, and the like.

Standard methods of peptide purification are employed to obtained isolated
peptide
compounds of the invention, including but not limited to various high-pressure
(or
performance) liquid chromatography (HPLC) and non-HPLC peptide isolation


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 48 -

protocols, such as size exclusion chromatography, ion exchange chromatography,
phase separation methods, electrophoretic separations, precipitation methods,
salting
in/out methods, immunochromatography, and/or other methods.

A preferred method of isolating peptide compounds useful in compositions and
methods of the invention employs reversed-phase HPLC using an alkylated silica
column such as C4-, C8- or C18-silica. A gradient mobile phase of increasing
organic
content is generally used to achieve purification, for example, acetonitrile
in an
aqueous buffer, usually containing a small amount of trifluoroacetic acid. Ion-
exchange
1o chromatography can also be used to separate peptide compounds based on
their charge.
The degree of purity of the peptide compound may be determined by various
methods,
including identification of a major large peak on HPLC. A peptide compound
that
produces a single peak that is at least 95% of the input material on an HPLC
column is
preferred. Even more preferable is a polypeptide that produces a single peak
that is at
least 97%, at least 98%, at least 99% or even 99.5% of the input material on
an HPLC
column.

To ensure that a peptide compound obtained using any of the techniques
described
above is the desired peptide compound for use in compositions and methods of
the
present invention, analysis of the compound's composition determined by any of
a
variety of analytical methods known in the art. Such composition analysis may
be
conducted using high resolution mass spectrometry to determine the molecular
weight
of the peptide. Alternatively, the amino acid content of a peptide can be
confirmed by
hydrolyzing the peptide in aqueous acid, and separating, identifying and
quantifying the
components of the mixture using HPLC, or an amino acid analyzer. Protein
sequenators, which sequentially degrade the peptide and identify the amino
acids in
order, may also be used to determine definitely the sequence of the peptide.
Since some
of the peptide compounds contain amino and/or carboxy terminal capping groups,
it
may be necessary to remove the capping group or the capped amino acid residue
prior
to a sequence analysis. Thin-layer chroinatographic methods may also be used
to
authenticate one or more constituent groups or residues of a desired peptide
compound.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-49-
Purity of a peptide compound may also be assessed by electrophoresing the
peptide
compound in a polyacrylamide gel followed by staining to detect protein
components
separated in the gel.

Assays to identify neuroprotective cornpounds
In addition to providing the exemplified neuroprotective peptide inhibitors of
AP-1
signaling, the present invention contemplates the identification of homologous
peptides
and small molecules which may be validated by the approaches disclosed herein.

io The identification of such homologs requires the establishment of (i) AP-1
signaling
inhibitory activity; and (ii) neuroprotective function as determined by their
ability to
reduce or inhibit neuronal cell death. Methods for conducting such assays are
clearly
described herein, in the accompanying examples.

Alternatively, or in addition to any one or more assays described in the
accompanying
examples, any one or more surrogate assays for determining (i) AP-1 signaling
inhibitory activity; and (ii) neuroprotective function of a homologous
compound may
be employed.

2o For example, AP-1 signaling inhibitors may be identified by their ability
to inhibit the
binding of AP-l to nucleic acid comprising AP-1 binding sites. In one
embodiment, an
AP-1 bZIP peptide is coated onto a microtitre plate and labelled
oligonucleotide (e.g.,
digoxigenin-labelled oligonucleotide) comprising AP-1 recognition site
sequences is
added to the microtitre plate in the presence or absence of a test compound.
Following
washing to remove unbound oligonucleotide, the amount of label bound to the AP-
1
peptide is determined. A compound that reduces the level of oligonucleotide
bound to
the peptide is considered to inhibit AP-1 signaling. Alternatively, or in
addition, AP-1
signaling inhibitory compounds are identified by their ability to inhibit
protein
interactions in the AP-1 signaling cascade, other than merely c-Jun
dimerization. For
3o example, a reverse hybrid assay can be employed to rescue cells in which a
test
compound inhibits or reduces an interaction between any of the following
proteins:


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-50-
Cdc42 and Racl, cdc42 and MLK3, Racl and MLK3, JNK and c-Jun, a MAP kinase
kinase kinase and JNK, JNK and JIP, or any of the proteins that interact to
form AP-1
e.g., ATF-2 or c-Fos. Reverse hybrid methods will be apparent to the skilled
artisan
and/or described in Watt et al. (USSN 09/227,652) or Erickson et al.
(W095/26400).

Homologs of the neuroprotective peptides described herein, including those
that have
been demonstrated to possess AP-1 signaling inhibitory activity by one or more
primary surrogate assays supra or by a method described in the accompanying
examples, may be determined by their ability to inhibit neuronal cell death
e.g.,
1o apoptosis and/or necrosis. For example, APOPTEST (available from
Immunotech)
stains cells early in apoptosis, and does not require fixation of the cell
sample (Martin
et al., 1994). This method utilizes an annexin V antibody to detect cell
membrane re-
configuration that is characteristic of cells undergoing apoptosis. Apoptotic
cells
stained in this manner can then sorted either by fluorescence activated cell
sorting
(FACS), ELISA or by adhesion and panning using immobilized annexin V
antibodies.
Alternatively, a terminal deoxynucleotidyl transferase-mediated biotinylated
UTP nick
end-labeling (TUNEL) assay is used to determine the level of cell death. The
TUNEL
assay uses the enzyme terminal deoxynucleotidyl transferase to label 3'-OH DNA
ends,
generated during apoptosis, with biotinylated nucleotides. The biotinylated
nucleotides
are then detected by using streptavidin conjugated to a detectable marker.
Kits for
TUNEL staining are available from, for example, Intergen Company, Purchase,
NY.
Alternatively, or in addition, an activated caspase, such as, for example,
Caspase 3 is
detected. Several caspases are effectors of apoptosis and, as a consequence,
are only
activated to significant levels in a cell undergoing programmed cell death.
Kits for
detection of an activated caspase are available from, for example, Promega
Corporation, Madison WI, USA. Such assays are useful for both
immunocytochemical
or flow cytometric analysis of cell death.



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-51-
Methods for detecting necrosis or determining the level of necrosis, e.g., in
a sample
comprising cells are known in the art and/or described, for example, in
Lemaire et al.,
Cell Death and Differentiation, 6: 813-820, 1999,

Therapeutic compositions
As will be apparent to the skilled artisan, peptides identified in the method
of the
present invention are useful as a therapeutic and/or prophylactic treatment of
a neuronal
disease and/or disorder, preferably those disorders associated with elevated
io extracellular glutamate leading to glutamate excitotoxicity and/or NMDA
excitotoxicity e.g., migraine, stroke, traumatic brain injury, epilepsy and
neurodegenerative disorders including Parkinson's Disease (PD), Alzheimer's
Disease
(AD) and Amyotrophic Lateral Sclerosis (ALS).

Accordingly, the present invention provides a method for preventing or
delaying
neuronal cell death in a subject comprising administering an AP-1 signaling
inhibitory
peptide of the invention according to any embodiment described herein or a
composition comprising said peptide to a subject in need of treatment e.g., a
subject
suffering from the disease and/or disorder or at risk of developing and/or
suffering from
the disease and/or disorder.

Clearly the present invention encompasses the use of an AP-1 signaling
inhibitory
peptide of the invention according to any embodiment described herein or a
derivative
or analog thereof in medicine. Additionally, the present invention encompasses
a
peptide identified by the present invention when used in medicine.

As will be apparent to the skilled artisan, the use of an AP-1 signaling
inhibitory
peptide of the invention according to any embodiment-described herein may
require the
peptide or analog be formulated into a composition for administration.

Preferably, the composition is a pharmaceutical composition.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-52-
To prepare pharmaceutical or sterile compositions including a peptide, peptide
analog,
peptide derivative or nucleic acid encoding the peptide, is mixed with a
pharmaceutically acceptable carrier or excipient. Compositions comprising a
therapeutic peptide or nucleic acid are prepared, for example, by mixing with
physiologically acceptable carriers, excipients, or stabilizers in the form
of, e.g.,
lyophilized powders, slurries, aqueous solutions, lotions, or suspensions
(see, e.g.,
Hardman, et al. (2001) Goodman and Gilman's The Pharmacological Basis of
Therapeutics, McGraw-Hill, New York, N.Y.; Gennaro (2000) Remington: The
1o Science and Practice of Pharmacy, Lippincott, Williams, and Wilkins, New
York,
N.Y.; Avis, et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral
Medications,
Marcel Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage
Forms:
Tablets, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical
Dosage
Forms: Disperse Systems, Marcel Dekker, NY; Weiner and Kotkoskie (2000)
Excipient
Toxicity and Safety, Marcel Dekker, Inc., New York, N.Y.).

Formulation of a pharmaceutical compound will vary according to the route of
administration selected (e.g., solution, emulsion, capsule). For solutions or
emulsions,
suitable carriers include, for example, aqueous or alcoholic/aqueous
solutions,
2o emulsions or suspensions, including saline and buffered media. Parenteral
vehicles can
include sodium chloride solution, Ringer's dextrose, dextrose and sodium
chloride,
lactated Ringer's or fixed oils, for instance. Intravenous vehicles can
include various
additives, preservatives, or fluid, nutrient or electrolyte replenishers and
the like (See,
generally, Remington's Pharmaceutical Sciences, 17th Edition, Mack Publishing
Co.,
Pa., 1985). For inhalation, the agent can be solubilized and loaded into a
suitable
dispenser for administration (e.g., an atomizer, nebulizer or pressurized
aerosol
dispenser).

Furthermore, where the active compound is a peptidyl compound, it can be
3o administered via in vivo expression of the recombinant protein. In vivo
expression can
be accomplished via somatic cell expression according to suitable methods
(see, e.g.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 53 -

U.S. Pat. No. 5,399,346). In this embodiment, nucleic acid encoding the
protein can be
incorporated into, a retroviral, adenoviral or other suitable vector
(preferably, a
replication deficient infectious vector) for delivery, or can be introduced
into a
transfected or transformed host cell capable of expressing the protein for
delivery. In
the latter embodiment, the cells can be implanted (alone or in a barrier
device), injected
or otherwise introduced in an amount effective to express the protein in a
therapeutically effective amount.

Selecting an administration regimen for a therapeutic composition depends on
several
io factors, including the serum or tissue turnover rate of the entity, the
level of symptoms,
the immunogenicity of the entity, and the accessibility of the target cells in
the
biological matrix. Preferably, an administration regimen maximizes the amount
of
therapeutic compound delivered to the patient consistent with an acceptable
level of
side effects. Accordingly, the amount of composition delivered depends in part
on the
particular entity and the severity of the condition being treated. Guidance in
selecting
appropriate doses of peptides are available (see, e.g., Milgrom, et al. New
Engl. J. Med.
341:1966-1973, 1999; Slamon, et al. New Engl. J. Med. 344:783-792, 2001;
Beniaminovitz, et al. New Engl. J. Med. 342:613-619, 2000; Ghosh, et al. New
Engl. J.
Med. 348:24-32, 2003; or Lipsky, et al. New Engl. J. Med. 343:1594-1602,
2000).


A peptide is provided, for example, by continuous infusion, or by doses at
intervals of,
e.g., one day, one week, or 1-7 times per week. Doses of a composition may be
provided intravenously, subcutaneously, topically, orally, nasally, rectally,
intramuscularly, intracerebrally, or by inhalation. A preferred dose protocol
is one
involving the maximal dose or dose frequency that avoids significant
undesirable side
effects. A total weekly dose depends on the type and activity of the compound
being
used. For example, such a dose is at least about 0.05 g/kg body weight, or at
least
about 0.2 g/kg, or at least about 0.5 g/kg, or at least about 1 g/kg, or at
least about
10 .g/kg, or at least about 100 g/kg, or at least about 0.2 mg/kg, or at
least about 1.0
mg/kg, or at least about 2.0 mg/kg, or at least about 10 mg/kg, or at least
about 25


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-54-
mg/kg, or at least about 50 mg/kg (see, e.g., Yang, et al. New Engl. J. Med.
349:427-
434, 2003; or Herold, et al. New Engl. J. Med. 346:1692-1698, 2002.

An effective amount of a peptide for a particular patient may vary depending
on factors
such as the condition being treated, the overall health of the patient, the
method route
and dose of administration and the severity of side affects, see, e.g.,
Maynard, et al.
(1996) A Handbook of SOPs for Good Clinical Practice, Interpharm Press, Boca
Raton,
Fla.; or Dent (2001) Good Laboratory and Good Clinical Practice, Urch Publ.,
London,
UK.
Determination of the appropriate dose is made by a clinician, e.g., using
parameters or
factors known or suspected in the art to affect tfeatment or predicted to
affect treatment.
Generally, the dose begins with an amount somewhat less than the optimum dose
and is
increased by small increments thereafter until the desired or optimum effect
is achieved
relative to any negative side effects. Important diagnostic measures include
those of
symptoms of the disease and/or disorder being treated. Preferably, a compound
that
will be used is derived from or adapted for use in the same species as the
subject
targeted for treatment, thereby minimizing a humoral response to the reagent.

2o An effective amount of therapeutic will decrease disease symptoms, for
example, as
described supra, typically by at least about 10%; usually by at least about
20%;
preferably at least about 30%; more preferably at least about 40%, and more
preferably
by at least about 50%.

The route of administration is preferably by, e.g., topical or cutaneous
application to an
open wound, or alternatively, by injection by intravenous, intraperitoneal,
intracerebral,
intramuscular, intraocular, intra-arterial, intracerebrospinal, intralesional,
intrathecal,
intra-arterial or pulmonary routes, or by local administration following a
craniotomy or
by sustained release or implant (see, e.g., Sidman et al. Biopolymers 22:547-
556, 1983;
3o Langer, et al. J. Biomed. Mater. Res. 15:167-277, 1981; Langer Chern. Tech.
12:98-
105, 1982; Epstein, et al. Proc. Natl. Acad. Sci. USA 82:3688-3692, 1985;
Hwang, et


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
- 55 -

al. Proc. Natl. Acad. Sci. USA 77:4030-4034, 1980; U.S. Pat. Nos. 6,350,466
and
6,316,024).

Formulations suitable for intracerebral, intrathecal, intra-arterial or
intracerebrospinal
injection or other injection directly to the central nervous system are
particularly
preferred, especially for conditions such as brain trauma or cerebral injuries
caused by
wounding e.g., motor vehicle accident etc. Formulations suitable for local
administration following a craniotomy are also particularly preferred.

io Preferred routes of administration of a neuroprotective peptide of the
invention are, for
example:
(i) intravenously, for example, in a 0.9% saline solution;
(ii) intrathecally, for example, the peptide composition is given after a
lumbar
puncture with a 18 G needle or after subsequent insertion of a extralumbal
catheter with
the tip in the intrathecal space;
(iii) by selective intra-arterial digital subtraction angiography, for
example, wherein
a microcatheter is inserted in the femoral artery and guided to the cerebral
arteries and
the peptide of the invention perfused into the area;
(iv) locally after craniotomy;
2o (v) by intracoronary delivery using catheter-based deliveries of
synthesized peptide
suspended in a suitable buffer e.g., such as saline which is injected locally
into the
coronary artery e.g., by injecting into the myocardium through the vessel
wall, using a
suitable local delivery catheter such as a 10 mm InfusaSleeve catheter (Local
Med,
Palo Alto, Calif.) loaded over a 3.0 mm x 20 mm angioplasty balloon, delivered
over a
0.014 inch angioplasty guide wire;
(vi) by intracoronary bolus infusion of peptide (or derivative) wherein the
peptide is
manually injected, for example, through an Ultrafuse-X dual lumen catheter
(SciMed,
Minneapolis, Minn.) or another suitable device into proximal orifices of
coronary
arteries;
(vii) by intramyocardial delivery of synthesized peptide or analog e.g., under
direct
vision following thoracotomy or using thoracoscope or via a catheter; or


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-56-
(viii) by application in a formulation comprising Gelfoam administered to a
lesion
e.g., in brain tissue.

Pericardial delivery of synthesized peptide or analog is typically
accomplished by
installation of the peptide-containing solution into the pericardial sac. The
pericardium
is accessed via a right atrial puncture, transthoracic puncture or via a
direct surgical
approach. Once the access is established, the peptide or analog is infused
into the
pericardial cavity and the catheter is withdrawn. Alternatively, the delivery
is
accomplished via the aid of slow-release polymers such as heparinal-alginate
or
Yo ethylene vinyl acetate (EVAc). In both cases, once the peptide or analog is
integrated
into the polymer, the desired amount of peptide/polymer is inserted under the
epicardial
fat or secured to the myocardial surface using, for example, sutures. In
addition, the
peptide/polymer composition can be positioned along the adventitial surface of
coronary vessels.

For administration of a peptide by a route that does not directly access the
central
nervous system, the peptide may have to cross the blood brain barrier. Methods
and
means for enabling a peptide to cross the blood brain barrier are known in the
art and/or
described, for example, in USSN20050142141. For example, a peptide of the
invention is conjugated to an agent that enables the peptide to cross the
blood brain
barrier (e.g., a Trojan horse). E.g., HIR MAb 83-14 is a murine MAb that binds
to the
human insulin receptor (HIR). This binding triggers transport across the BBB
of MAb
83-14 (Pardridge et al, Pharm., Res. 12: 807-816, 1995), and any drug or gene
payload
attached to the MAb (Wu et aL, J. Clin. Invest., 100: 1804-1812, 1997).
The use of molecular Trojan horses to ferry drugs or genes across the blood
brain
barrier is described in U.S. Pat. Nos. 4,801,575 and 6,372,250. The linking of
drugs to
MAb transport vectors is facilitated with use of avidin-biotin technology. In
this
approach, the drug or protein therapeutic is monobiotinylated and bound to a
conjugate
of the antibody vector and avidin or streptavidin. The use of avidin-biotin
technology to
facilitate linking of drugs to antibody-based transport vectors is described
in U.S. Pat.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
-57-
No. 6,287,792. Fusion proteins have also been used where a drug is genetically
fused to
the MAb transport vector.

In a preferred embodiment, a therapeutic peptide described herein is
administered to a
subject when the subject is suffering from or has recently suffered from an
ischemic
event (e.g., a stroke) or nerve trauma or trauma to the central nervous
system. Such
timing of administration is useful for, for example, reducing the effect of
reperfusion
following an ischemic event. The peptide may also be administered to a subject
when
the subject is at risk of experiencing a reperfusion injury following an
ischemic event.
io
The present invention is further described with reference to the following non-
limiting
examples.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
58
EXAMPLE 1
Isolation of AP-1 complex formation inhibitory PhylomerTM peptides
PhylomerT`x peptide Library Construction
Genomic DNA from 15 different sequenced bacterial genomes (Aquifex aeolicus,
Bacillus subtilis, Bordetella pertussis, Borrelia burgdorferi, Chlamydia
trachomatis,
Escherichia coli K12, Haemophilus influenzae, Helicobacter pylori,
Methanobacterium
thermoautotrophicu, Methanococcus jannashii, Neisseria meningitides,
Pyrococcus
horikoshi, Pseudomonas aeruginosa, Synechocystis PCC 6803, Thermoplasma
volcanicum) was obtained. This DNA was used as the template for random
amplification using random primers containing a FLAG-tag, designated BGFN6 and
BGFN9, as follows:

BGFN6:
5'-GACTACAAGGACGACGACGACAAGGCTTATCAATCAATCANNNNNN-3;
BGFN9:
5'-GACTACAAGGACGACGACGACAAGGCTTATCAATCAATC -3'
The following protocol was employed:
Amplification Round 1: 3.33 M Klenow primer, 1X Klenow buffer, 200 M dNTPs,
Klenow, PEG (8500) in total volume of 30 1. Mix primer, DNA, and water; boil
for 3-
5 min, snap cool on ice and then transfer to tube containing the other
reagents. Incubate
15 C for 30 mins, RT for 2 hours, then 37 C for 15 min.
Amplification Round 2: Boil tube 5 min, snap cool, add 0.5 l Klenow enzyme
and then
incubate as in step 2.
Amplification Round 3: Boil tube 5 min, snap cool, add: 4 1 BGF-F9 primer (25
M),
l l lOX buffer, 3 1 dNTPs (2mM), 0.5 1 Klenow, 1.5 1 water. Incubate 15 C for
30
min, RT for 2 hours, 37 C for 15 min.
Amplification Round 4: Boil tube 5 min, snap cool, add 0.5 1 Klenow enzyme and
then
incubate as in Round 3 and products purified using Amplicon spin columns.

Cloning sites were added by conventional PCR primed from the tag sequence with
the
following primers:


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
59
BGF-F5:
5'-GAGAGgaattcAGGTCAGACTACAAGGACGACGACGACAAG-3';
BGF-R6-Acc651:
5'-GAGAGggtaccAGGTCAGACTACAAGGACGACGACGACAAG-3'.

The amplified sequences were digested with EcoRIlAcc651, and cloned into the
vector
pYTB3, a TRPI expressing, 2-micron origin vector allowing constitutive
expression of
inserts from the ADH1 promoter. About 5 x 106 colony forming units (CFUs) were
formed from the primary transformation and harvested from the plates. DNA was
prepared from harvested cells using standard procedures.

Reverse-2-hybr id Screening
The yeast two hybrid system described by Vidal.M., In: The Yeast Two Hybrid
System
(eds. P.Bartel and S. Fields), Oxford University Press, New York (1997), was
modified
to add another counter selectable marker (CYH2) and to allow flexible
titration of
stringency by adjustment of sugar concentrations in the screening media.

As an example to demonstrate the ability of peptides to inhibit AP-1 complex
formation, inhibition of c-Jun dimerization was tested. Partial fragments of c-
Jun i.e.,
JunZ (774-927 bp) and Jun1 (558-1002 bp), were cloned into yeast two-hybrid
vectors
pDD (a kanamycin-resistant variant of pGilda bait vector) and pJFK (pYesTrp
prey
vector (Invitrogen) modified by replacing the TRPI yeast selection gene with
HIS5),
respectively, before co-transforming into Saccharomyces cerevisiae strain
PRT480
(MATa, his3, trpl, ura3, 4 LexA-LEU2, lys2::3 clop-LYS2, CANR , CYH2R ,
ade2::2
LexA-CYH2-ZEO, his5::2 LexA-URA3-G418) using a standard lithium-acetate based
chemical transformation protocol.

The PhylomerTM peptide library was transformed into S. cerevisiae strain PRT51
(MATa, his3, trpl, ura3, 6 LexA-LEU2, lys2::3 clop-LYS2, CYH2R , ade2::G418-
pZero-ade2, met15::Zeo-pBLUE-met15, his5::hygro), using a high-efficiency
lithium


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
acetate-based chemical transformation protocol, slightly modified to maximize
the
number of transformants.

Bait/prey plasmid containing PRT480 haploids (108 cells) were mated with the
5 PhylomerTM library (107 c.f.u.) according to a liquid mating protocol for
pretransformed libraries (Clontech, USA), and plated to HW- minimal media
(minimal
media lacking histidine and tryptophan) to select for diploids. These plates
were
scraped after 2 days incubation at 30 C, and the cells were washed,
resuspended 1:1
(v/v) in yeast freezing solution (65% v/v glycerol, 0.1M MgSO4, 25mM Tris-Cl
pH
10 8.0), and frozen at -80'C in lml aliquots.

To select peptides that block formation of an AP-1 complex requiring the
interaction
between JUN1 and JunZ, about 1.5 x 107 c.f.u Jun/PhylomerTm diploids were
thawed
and outgrown overnight in HW- to achieve log-phase growth. The following day,
4 x
15 107 cells were plated onto counter-selective media: HWU (lacking histidine,
tryptophan and uracil), containing supplements of 0.02% galactose (gal), 2%
raffinose
(raff), 0.2 g/ml uracil, 0.06% (w/v) 5-Fluoroorotic acid (FOA), 5 g/ml
cycloheximide.
These plates were incubated for 7 days, then colonies were picked to HWU-
0.02% gal,
2% raff, and then to HWL- (lacking histidine, tryptophan and leucine) 0.02%
gal, 2%
20 raff to confirm blocking phenotype.

From an initial reverse-two-hybrid screen of approximately 300,000 clones
expressed
from a third vector (pYTB3), 95 primary transformants were identified that
allowed
yeast survival on media for counter-selection against an AP-1 complex
formation viz.
25 c-Jun dimerization. These included transformants comprising the clones
designated
PYC19 [DNA sequence set forth in SEQ ID NO: 26; encoded amino acid sequence
set
forth in SEQ ID NO: 27 (with FLAG epitope) and 28 (without FLAG epitope)];
PYC35 [DNA sequence set forth in SEQ ID NO: 34; encoded amino acid sequence
set
forth in SEQ ID NO: 35 (with FLAG epitope) and 36 (without FLAG epitope)];
PYC36
30 [DNA sequence set forth in SEQ ID NO: 43; encoded amino acid sequence set
forth in
SEQ ID NO: 44 (with FLAG epitope) and 45 (without FLAG epitope)]; PYC38/39


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
61
[DNA sequence set forth in SEQ ID NO: 52; encoded amino acid sequence set
forth in
SEQ ID NO: 53 (with FLAG epitope) and 54 (without FLAG epitope)]; and PYC41
[DNA sequence set forth in SEQ ID NO: 60; encoded amino acid sequence set
forth in
SEQ ID NO: 61 (with FLAG epitope) and 62 (without FLAG epitope)].

Subsequently, the individual plasmid clones were rescued from the yeast and re-

transformed to confirm the disruption phenotype; 63% of the primary "hits",
including
PYC 19, PYC35, PYC26, PYC38/39 and PYC41, were positive in two assays for
recapitulation of the phenotype.

EXAMPLE 2
PhylomerTM peptides that decrease AP-1 controlled reporter gene expression
PhylomerTM peptides that inhibit an AP-1 complex formation as described in
Example
1 were tested for their ability to decrease AP-1 controlled reporter
expression (since Jun
dimers are AP-1 moieties). A transient transfection assay in which the
expression of
each clone was plasmid driven was employed, thereby avoiding the synthesis of
large
numbers of peptides.

Luciferase reporter gene assay of,4.P-1 dependent transcriptional activity
The K562 cell line was stably-transfected with the AP-1 luciferase reporter
(AP1-Luc)
of the Mercury Pathway Profiling kit (Clontech, U.S.A.), and the clonal cell
line
K562/APl-Luc was established. In 6-well tissue culture plate format, K562/AP1-
Luc
cells were transfected with either pcDNA3 control, pcDNA3-JunDN (a dominant
negative mutant of c-Jun), or pcDNA3-peptide, using Lipofectamine2000 (Life
Technologies) according to manufacturer's instructions. Transfection reactions
were
incubated for 48 hours, cells collected and protein lysates extracted for
luciferase assay
according to Mercury Pathway Profiling kit and associated protocols.

3o Luciferase assays were performed in independent triplicates, and results
for each
peptide subjected to statistical analysis (SPSS software package) to determine
if they


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
62
were different to JunDN (positive control for AP-1 inhibition) or pcDNA-3
(negative
control for AP-1 inhibition).

All AP-1 luciferase values were normalised to the renilla expression from a
cotransfection control. Under these experimental conditions, approximately 30%
of
PhylomerTM peptides decreased AP-1 reporter activity by at least 50% when
compared
to the vector-only control (i.e., pcDNA3 vector). More particularly, the
peptides
designated PYC35, PYC36, PYC38/39 and PYC41 reduced AP-1 reporter activity to
approximately 35%, 44%, 47% and 48%, respectively of the level observed for
the
control sample. These data demonstrate that the hit-rate from the c-Jun screen
for
functional blockers of AP-1 activity was high i.e., exceeding 1:13,000,
suggesting that
PhylomerTM peptide libraries provide a rich source of AP-1 inhibitors.

EXAMPLE 3
Peptides that inhibit AP-1 signaling are neuroprotective following glutamate
excitotoxicity in primary cortical neuronal cultures

Materials &Methods
1. Peptides
Peptides used in this example comprise the amino acid sequences shown in the
accompanying Sequence Listing. The peptides were designated as shown in Table
1.
TABLE 1

Peptide Name Description Sequence SEQ ID
NO.
PYC19L Phylomer M core sequence Leu lie Asn Gln Ser Tyr Ala Tyr Pro Tyr Ile
Tyr SEQ ID NO:
28
PYC19D Retroinverted fonn of PYC19L8 Tyr Ile Tyr Pro Tyr Ala Tyr Ser Gln Asn
Ile Leu Gly SEQ ID NO:
29
PYC19L-TAT PYC19L with an N-terminal TAT Gly Arg Lys Lys Arg Arg Gin Arg Arg
Arg Gly Leu Ile SEQ ID NO:
Asn Gin Ser Tyr Ala Tyr Pro Tyr Ile Tyr 30
PYC19D-TAT Retroinverted PYC19L-TAT Tyr Ile Tyr Pro Tyr Ala Tyr Ser Gln Asn
Ile Leu Gly. SEQ ID NO:
Ar A-rg Arg Gln Ar Ar Lys Lys Arg Gly 31
PYC19L-FM PYC19L with C-terminal Kaposi Leu lie Asn Gln Ser Tyr Ala Tyr Pro
Tyr Ile Tyr Gly SEQ ID NO:
FGF protein transduction domain Ala Ala Val Leu Leu Pro Val Leu Len Ala Ala
Pro 32
PYC19D-FM Retroinverted form of PYC19L- Pro Ala Ala Leu Leu Val Pro Leu Leu
Val Ala Ala Gly SEQ ID NO:
FMb Tyr Ile Tyr Pro Tyr Ala Tyr Ser Gln Asn Ile Leu 33
PYC35L Phylomer' core sequence Ala Tyr Gin Ser Ile Arg Ser Gly Gly Ile Glu Ser
Ser Ser SEQ ID NO:
L s Ar Glu Ar 36
PYC35D Retroinverted form of PYC35La Arg Glu Arg Lys Ser Ser Ser Glu Ile Gly
Gly Ser Arg SEQ ID NO:
Ile Ser Gin Tyr Ala Gly 37


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
63
PYC35L-TAT PYC35L witli an N-terminal TAT Gly Arg Lys Lys Arg Arg Gln Arg Arg
Arg Gly Ala SEQ ID NO:
Tyr Gln Ser Ile Arg Ser Gly Gly Ile Glu Ser Ser Ser Lys 38
Arg Glu Arg
PYC35D-TAT retroinverted foxm of PYC35L- Arg Glu Arg Lys Ser Ser Ser Glu Ile
Gly Gly Ser Arg SEQ ID NO:
TAT' Ile Ser Gln Tyr Ala Gly Arg Arg Arg Gln Arg Arg Lys 39
Lys Ar Gly
PYC35L-FM PYC35L with C-terminal Kaposi Ala Tyr Gln Ser Ile Arg Ser Gly Gly
Ile Glu Ser Ser Ser SEQ ID NO:
FGF protein transduction domain Lys Arg Glu Arg Gly Ala Ala Val Leu Leu Pro
Val Leu 40
Leu Ala Ala Pro
PYC35D-FM retroinverted form of PYC35L- Pro Ala Ala Leu Leu Val Pro Leu Leu
Val Ala Ala Gly SEQ ID NO:
FM' Arg Glu Arg Lys Ser Ser Ser Glu Ile Gly Gly Ser Arg 41
Ile Ser Gln T Ala
PYC35DScram- retroinverted peptide comprising Lys Ile Glu Arg Ser Glu Gly Ile
Ser Gin Ser Ala Arg SEQ ID NO:
TAT scrambled PYC35D peptide and Ser Arg Gly Tyr Ser Gly Arg Arg Arg Gln Arg
Arg Lys 42
retroinverted TATb Lys Arg Gly
PYC36L PhylomerTM core sequence Gly Leu Gln Gly Arg Arg Arg Gln Gly Tyr Gln
Ser Ile SEQ ID NO:
Lys Pro 45
PYC36D retroinverted form of PYC36L Pro Lys Ile Ser Gln Tyr Gly Gln Arg Arg
Arg Gly Gln SEQ ID NO:
Leu Gl 46
PYC36L-TAT PYC36L with an N-terminal TAT Gly Arg Lys Lys Arg Arg. Gin Arg Arg
Arg Gly Gly SEQ ID NO:
Leu Gln Gly Arg Arg Arg Gln Gly Tyr Gln Ser Ile Lys 47
Pro
PYC36D-TAT retroinverted form of PYC36L- Pro Lys Ile Ser Gin Tyr Gly Gln Arg
Arg Arg GIy Gln SEQ ID NO:
TATb Leu Gly Gly Arg Arg Arg Gln Arg Arg Lys Lys Arg 48
Gly
PYC36L-FM PYC36L with C-terminal Kaposi Gly Leu Gln Gly Arg Arg Arg Gln Gly
Tyr Gln Ser Ile SEQ ID NO:
FGF protein transduction domain) Lys Pro Gly Ala Ala Val Leu Leu Pro Val Leu
Leu Ala 49
Ala Pro
PYC36D-FM retroinverted form of PYC36L- Pro Ala Ala Leu Leu Val Pro Leu Leu
Val Ala Ala Gly SEQ ID NO:
FM' Pro Lys Ile Ser Gln Tyr Gly Gln Arg Arg Arg Gly Ghi 50
Leu Gly
PYC36D Scram- retroinverted peptide comprising Lys Arg Arg Gly Gly Ile Leu Arg
Tyr Gly Gln Pro Gln SEQ ID NO:
TAT scrambled PYC36D peptide and Ser Gln Gly Arg Arg Arg Gln Arg Arg Lys Lys
Arg 51
retroinverted TAT protein Gly
transduction domainb
PYC38/39L PhylomerTM core sequence Gly Leu Gln Gly Arg Arg Gln Pro Gly Gln Gln
Pro Gln SEQ ID NO:
GI Arg Trp Ser Gl Ar Ala Leu Pro Ala His Arg 54
PYC38/39D retroinverted form of PYC38/38L Arg His Ala Pro Leu Ala Arg Gly Ser
Trp Arg Gly Gln SEQ ID NO:
Pro Gin Gln Gly Pro Gln Arg Arg Gly Gln Leu Gly 55
PYC38/39L-TAT PYC38/39L with N-terminal TAT Gly Arg Lys Lys Arg Arg Gln Arg
Arg Arg Gly Gly SEQ ID NO:
Leu Gln Gly Arg Arg Gln Pro Gly Gln Gln Pro Gln Gly 56
Arg Trp Ser Gly Arg Ala Leu Pro Ala His Arg
PYC38/39D-TAT retroinverted PYC3 8/3 9L-TAT Arg His Ala Pro Leu Ala Arg Gly
Ser Trp Arg Gly Gln SEQ ID NO:
Pro Gln Gin Gly Pro Gln Arg Arg Gly Gln Leu Gly Gly 57
Arg Arg Arg Gln Ar Ar Lys Lys Arg Gly
PYC38/39L-FM PYC38/39L with C-terminal Gly Leu Gln Gly Arg Arg Gln Pro Gly Gln
Gln Pro Gln SEQ ID NO:
Kaposi FGF protein transduction Gly Arg Trp Ser Gly Arg Ala Leu Pro Ala His
Arg Gly 58
domain Ala Ala Val Leu Leu Pro Val Leu Leu Ala Ala Pro
PYC38/39D-FM retroinverted PYC38/39L-FM Pro Ala Ala Leu Leu Val Pro Leu Leu
Val Ala Ala Gly SEQ ID NO:
Arg His Ala Pro Leu Ala Arg Gly Ser Trp Arg Gly Gln 59
Pro Gln Gin Gly Pro Gln Ar Ar Gly Gln Leu Gly
PYC41L Phylomer core sequence Val Ser Ile Asn Gln Glu His His Arg Leu Leu Pro
Leu SEQ ID NO:
62
PYC41D retroinverted PYC41L Leu Pro Leu Leu Arg His His Glu Gln Asn Ile Ser
Val SEQ ID NO:
G1 63
PYC41L-TAT PYC41L with N-terminal TAT Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg
Gly Val SEQ ID NO:
Ser Ile Asn Gln Glu His His Ar Leu Leu Pro Leu 64
PYC41D-TAT retroinverted PYC4IL-TAT Leu Pro Leu Leu Arg His His Glu Gln Asn
Ile Ser Val SEQ ID NO:
Gl Ar Ar Ar Gln Ar Ar L s L s Ar GI 65
PYC41L-FM PYC41 with C-terminal Kaposi Val Ser Ile Asn Gln Glu His His Arg Leu
Leu Pro Leu SEQ ID NO:
FGF protein transduction domain GIy Ala Ala Val Leu Leu Pro Val Leu Leu Ala
Ala Pro 66
PYC41D-FM retroinverted PYC41L-FM Pro Ala Ala Leu Leu Val Pro Leu Leu Val Ala
Ala Gly SEQ ID NO:
Leu Pro Leu Leu Arg His His Glu Gln Asn Ile Ser Val 67
JNICl-1D-TAT retroinverted JNK1-I-TAT Thr Asp Gln Ser Arg Pro Val Gln Pro Phe
Leu Asn Leu SEQ ID NO:
Thr Thr Pro Arg Lys Pro Arg Pro Pro Arg Arg Arg Gln 68
Ar Arg Lys Lys Arg Gly
a, all amino acids other than glycine are D-amino acids with a C-terminal
glycine linker added
b, alI amino acids other than glycine are D-amino acids


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
64
All peptides are synthesized using standard procedures and HPLC-purified,
e.g., by
Mimotopes Pty Ltd (Australia), or GenScript Corporation (USA), or Auspep
(Australia). All peptides were prepared as 100x stocks (500 M) in normal
saline.

The peptide JNKI-1 D-TAT served as a positive control. Peptides PYC35D Scram-
TAT and PYC36D Scram-TAT served as negative controls, and to determine whether
or not the integrity of the retroinverted PhylomerTM peptide sequence was
necessary for
proficiency in attenuating neuronal cell death.

2. Primarticortical neuronal cultures
Cortical neuronal cultures were established as described by Meloni et al.,
Neuroscience
108, 17-26, 2001. Briefly, cortical tissue from E18-E19 Sprague-Dawley rats
was
dissociated in Dulbelcco's Modified Eagle Medium (DMEM; Invitrogen, Australia)
supplemented with 1.3mM L-cysteine, 0.9mM NaHCO3, 10 units/ml papain (Sigma,
USA) and 50 units/ml DNase (Sigma) and washed in cold DMEM/10% (v/v) horse
serum. Neurons were resuspended in Neurobasal (NB; Invitrogen) containing 2%
B27
supplement (B27; Invitrogen). Before seeding, culture vessels comprising
either a 96-
well plastic plate or 24-well plastic plate with 13mm glass coverslips
(ProSciTech,
Australia), were coated with poly-D-lysine (50 g/mL; 70 - 150K; Sigma) and
incubated overnight at room temperature. The poly-D-lysine was removed and
replaced with NB (containing 2% B27; 4% fetal bovine serum; 1% horse serum;
62.5 M glutamate; 25 .M 2-mercaptoethanol; and 30 g/mL streptomycin and 30
g/mL penicillin). Neurons were plated to obtain approximately 10,000 viable
neurons
for each well of a 96-well plate, or 200,000 viable neurons per well of a 24-
well plate,
on day in vitro (DIV) 9. Neuronal cultures were maintained in a COZ incubator
(5%
C02, 95% air balance, 98% humidity) at 37 C. On DIV 4, one third of the
culture
medium was removed and replaced with fresh NB/2% B27 containing the mitotic
inhibitor, cytosine arabinofuranoside (Sigma) at 1 M concentration. On DIV 8,
one
half of the culture medium was replaced with NB/2% B27. Cultures were used on
DIV
11 or 12, and consisted of >95% neurons.


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
3. Glutamate excitotoxicity
To induce glutamate excitotoxicity in the cortical neuronal cultures (96-well
plate
format), 50 l of conditioned media containing 200 M glutamate was added to
culture
5 wells containing 50 l conditioned media (100 M final glutamate
concentration).
Cultures were incubated at 37 C in a COz incubator for 5 minutes, after which
time the
media was replaced with 100g1 of 50% NB/2% N2 and 50% balance salt solution
(NB/N2:BSS).

10 To determine the efficacy of a peptide in preventing or delaying glutamate
excitotoxicity, peptides were added to wells at a suitable time prior to
incubation of
neurons in the presence of glutamate, e.g., about 15 minutes prior to
incubation in
glutamate or post-glutamate exposure.

15 For determining the time course over which any peptide was effective,
peptides were
added either prior to or post-glutamate exposure.

To determine calcium influx, peptides were added both prior to and post-
glutamate
exposure.

A non-peptide positive control, consisting of 5 M concentration of the
glutamate
receptor inhibitors MK801/5 M 6-cyano-7-nitroquinoxaline (MK801/CNQX), was
used in a similar manner to peptides, either prior to or post-glutamate
exposure.

Untreated and glutamate-treated control samples received media additions with
and
without glutamate respectively.

4. Neuronal viability and statistical analysi
Eighteen hours after glutamate exposure, neuronal cultures were examined by
light
microscopy for qualitative assessment of neuronal damage. Neuronal viability
was
quantitatively measured by 3-(4,5,dimethyliazol-2-yl)-5-(3-carboxymethoxy-
phenyl)-2-


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
66
(4-sulfophenyl)-2H-tetrazolium salt (MTS) assay (Promega, Australia). The MTS
assay
measures the mitochondrial conversion of the tetrazolium salt to a water-
soluble brown
formazan salt, which is detected spectrophotometrically at 495nm. MTS
absorbance
data was converted to reflect proportional cell viability relative to both the
untreated
and glutamate treated controls. Viability data was analysed by ANOVA, followed
by
post-hoc Fischer's PLSD test, with p<0.05% values considered statistically
significant.
All assays were performed with quadruplicate sister neuronal cultures repeated
a
minimum of four times independently.

5. Neuronal intracellular calcium levels followingglutamate exposure
Cortical cultures (24-well plate) were loaded with the Ca2+ indicator dye Fura-
2-AM
(1 M) in 300 l NB/N2:BSS, 0.3% pluronic F-127, for 45 minutes at 37 C. The
loading media was then exchanged for 300 1 of fresh NB/N2:BSS containing 5 M
peptide or MK801/CNQX and incubated for 20 minutes at 37 C. Control cultures
received 300 1 of NB/N2:BSS only. Coverslips were transferred to a microscope
recording chamber of the microscope in 900 .l physiological rat saline (PRS)
only, or
containing peptide (5 M) or MK801/CNQX and incubated for a further 10 minutes
at
room temperature. A diaphragm device located on the microscope optically
isolated a
group of five to. seven neurons for each measurement. Intracellular Ca2+
levels were
recorded for 30 seconds before the addition of 100 l of 1mM glutamate (100 M
final
concentration), and for a further 210 seconds after the addition. Measurements
of Ca2+
were performed using an inverted epifluorescence microscope (Nikon TE2000,
Japan)
connected to a spectrophotometer (Cairn, UK). The ratio (R) of fluorescence
emission
(emission wave-length: 510nm) at 340- and 380-nm excitation (F340/F380), was
collected at 10 Hz, stored and analyzed using the Cairn software package
(Cairn).

Results
Screening of PhylomerTM peptides in the glutamate excitotoxicity model
indicated that
the peptides designated PYC19L-TAT, PYC35L-TAT, PYC36L-TAT, PYC38/39L-
3o TAT and PYC41L-TAT display neuroprotective activity (Figure 1). Following
glutamate excitotoxicity, the peptides increased neuronal viability from 2-5%
basal


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
67
level, to 32% for PYC19L-TAT, 42% for PYC35L-TAT, 64% PYC36L-TAT, 69% for
PYC38/39L-TAT, and 38% for PYC41L-TAT. The positive peptide control JNKI-1 D-
TAT increased neuronal viability to 75%. In contrast, TAT peptide alone or
scrambled
PhymolerTM peptides i.e., PYC35D Scram-TAT or PYC36D Scram-TAT, failed to
provide neuroprotection in the glutamate model (Figure 2).

Neuronal viability correlated with morphological assessment observed by light
microscopy (Figure 3). At one hour post-glutamate exposure, neurons in
untreated
cultures and cultures treated with control peptides (TAT, PYC35D Scram-TAT,
PYC36D Scram-TAT, PYC35D, PYC36D) displayed cellular rounding, and, by 18
hours post-exposure, few neurons (2-5%) appeared intact and viable. In
contrast, most
neurons in cultures treated with neuroprotective AP-1 signaling inhibitory
peptides
e.g., PYC19L-TAT, PYC35L-TAT, PYC36L-TAT, PYC38/39L-TAT, PYC41L-TAT).
Or with the positive control peptide designated JNKI-1 D-TAT, or with
glutamate
receptor inhibitor, did not begin to round and survived glutamate
excitotoxicity.

The neuroprotective PhylomerTm peptides also increased neuronal survival in a
dose
dependent fashion following glutamate excitotoxicity (Figure 4). For example,
an
extracellular concentration of l M PYC36L-TAT provided a significant level of
neuroprotection, and the same extracellular concentration of the peptides
PYC19L-
TAT, PYC38/39L-TAT and PYC41L-TAT provided a neuroprotective trend. At an
extracellular concentration of 2 M, peptides PYC38/39L-TAT and PYC41L-TAT
provided significant neuroprotection, and peptides PYC19L-TAT and PYC35L-TAT
exhibited a neuroprotective trend. At extracellular concentrations of 5 M and
10 M,
all five peptides provided significant neuroprotection.

Dose response curves showing the neuroprotective efficacies of L- and D-
isoforms of
these PhylomerTM peptides demonstrate that the D-isoforms are more potent
(Figures
4a-e).



CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
68
For example, an extracellular concentration of 1 M PYC 19D-TAT or PYC 19L-TAT
provides significant neuroprotection (Figure 4a), however at higher
concentrations,
PYC19D-TAT clearly provides enhanced neuroprotection. In addition, at 5 M,
neuronal survival increased from 32% using PYC19L-TAT to 68% using the
retroinverted form PYC19D-TAT.

Similar results were also obtained for PYC35-TAT peptide (Figure 4b), wherein
500nM PYC35D-TAT provided significant neuroprotection, compared to a
requirement
for 2 M or greater concentration of PYC35L-TAT to provide significant
neuroprotection; and neuronal survival at 5 M extracellular peptide
concentration
increasing from 42% using PYC35L-TAT to 59% when using PYC35D-TAT.
Similarly, at 5 M extracellular peptide concentration, neuronal survival
increased from
64% using PYC36L-TAT to 82% when using PYC36D-TAT (Figure 4c). In the case
of peptides PYC36L-TAT and PYC36D-TAT, whilst significant neuroprotection was
achieved at 1 M extracellular concentration for both isoforms, a
significantly greater
level of viability was achieved using the D-isoform, i.e., 44% for PYC36D-TAT
cf.
30% for PYC36L-TAT. At lower extracellular peptide concentrations e.g., 0.1 M
and
0.5 M, the D-isoform also displayed significant neuroprotection compared to
the L-
isoform.

The administration of 2 M PYC38/39L-TAT, PYC41L-TAT, PYC38/39D-TAT or
PYC41D-TAT achieved an initial significant neuroprotection (Figures 4d,e); and
at
5 M extracellular peptide concentration, neuronal viability increased from 69%
using
PYC38/39L-TAT to 83% using PYC38/39D-TAT (Figure 4d), however did not vary
significantly for the peptides PYC41D-TAT and PYC41L-TAT (40% cf. 38%; Figure
4e).

These data compare favorably with the neuronal protection conferred for the
JNKI-ID-
3o TAT peptide, which provided significant neuroprotection at an extracellular
concentration of 2 M or greater, to a maximum neuronal viability of 89% at 5
M


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
69
extracellular concentration i.e., comparable to efficacy of 5 M PYC36D-TAT. In
particular, at 1 M concentration, the PhylomerTM peptides PYC19D-TAT, PYC36L-
TAT, PYC36D-TAT and PYC38/39D-TAT at least provided greater neuroprotection
than JNKI-ID-TAT; and at 2 M concentrations the PhylomerTM peptide PYC36D-
TAT provided greater neuroprotection than JNKI-ID-TAT. Moreover, 500nM
PYC35D-TAT provides significant (p <0.0242) neuroprotection (Figure 4b), and
100
nM PYC36D-TAT provides significant (p <0.0196) protection (Figure 4c),
compared
to the same concentration of JNK1-ID-TAT. In fact, the peptide JNKl-1D-TAT
only
exhibits significant (p <0.0077) neuroprotection at micromolar concentrations
under
1o these conditions.

The efficacy of the FM and TAT transduction domains, synthesised with both L-
and
D-isoforms of peptides PYC35 and PYC36, were also compared (Figure 5). Neither
L-
nor D-isoforms of PYC35-FM and PYC36-FM provided significant neuroprotection
following glutamate excitotoxicity, compared to PYC35D-TAT and PYC36D-TAT.
Peptides were also administered over a time course, for peptides PYC35D-TAT
and
PYC36D-TAT. In particular, these peptides were added to neuronal cultures
prior to,
immediately post (0 min), or at 15, 30, 45 or 60 minutes after glutamate
exposure
(Figure 6). Data indicate that administration of either PYC35D-TAT or PYC36D-
TAT
prior to glutamate exposure was neuroprotective.

Peptide treated and control cultures were also assessed for neuronal
intracellular Ca2+
influx following glutamate exposure. Cultures treated with peptides PYC35D-TAT
or
PYC36D-TAT and exposed to glutamate did not block neuronal Ca2+ entry. Rather,
there was a rapid influx of Ca2+, with measured amplitudes of 361 and 442
respectively
(Figure 7). Similarly, neurons in control cultures exposed to glutamate also
underwent
rapid increases in intracellular Ca2+ with an amplitude of 496. In contrast,
neuronal
cultures treated with glutamate blockers MK801/CNQX exposed to glutamate
inhibited
intracellular Ca2+ influx with a nominal amplitude of 26 (Figure 7).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
In summary, this study has validated five AP-1 signaling inhibitory peptides
that block
neuronal death following glutamate excitotoxicity. As both c-Jun activation
and
glutamate excitotoxicity are linked to neurodegenerative disorders, cerebral
ischemia,
traumatic brain injury, epilepsy, Parkinson's Disease, Alzheimer's Disease and
5 Amyotrophic Lateral Sclerosis (ALS), these peptides are these peptides have
utility for
the development of compositions for the treatment of such neurological
disorders. In
vivo studies will validate these peptides as neuroprotectants in animal models
for
various neurodegenerative disorders.

10 EXAMPLE 4
Peptides that inhibit AP-1 signaling are neuroprotective following N-methyl -D-

aspartate (NMDA) excitotoxicity in primary cortical neuronal cultures
NMDA-induced excitotoxicity specifically targets the NMDA glutamate receptor
to
1.5 induce excitotoxicity and induces an acute form of cell death in neuronal
cultures.
Materials & Methods
1. Peptides

Peptides used to determine rescue of neurons from NMDA-induced excitotoxicity
were
20 PYC35D-TAT and PYC36D-TAT as shown in Table 1.

2. NMDA excitotoxicity
The procedure for determining NMDA excitotoxicity in neuron culture is similar
to the
procedure for determining glutamate excitotoxicity (Example 3), however a
final
25 NMDA concentration of 200 M is used, and the insult time is for 15
minutes.

Results
The neuroprotective PhylomerTM peptides PYC35D-TAT and PYC36D-TAT, which
have been shown herein to rescue neurons from glutamate excitotoxicity, also
protect
30 cortical neurons against NMDA-induced excitotoxicity (Figure 8; p <0.0001
in both
cases).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
71
EXAMPLE 5
Peptides that inhibit AP-1 signaling reduce cell death
in an in vitro ischemia cell model

The ability of retroinverted Phylomerrm peptides to protect cultured cortical
neurons in
an in vitro model of ischemia known as Oxygen Glucose Deprivation (OGD). This
model involves inducing an energy crisis in neuronal cultures by transiently-
depriving
the neurons of oxygen and glucose for 35 minutes, which are essential for
normal
cellular metabolism and the main energy substrates that are depleted during
cerebral
ischemia.

Materials and methods
1. Determination of ischemia in vitro
Ischemia in vitro is determined in glass wells, because plastic wells store
oxygen, by
removing media from neuronal cultures in glass wells and washing in 315 l
balanced
salt solution (BSS; mM: 116 NaCl, 5.4 KCI, 1.8 CaC12, 0.8 MgSO4, 1 NaH2PO4; pH
7.3) and re-adding 50 1 of BSS containing 25 mM 2-deoxy-D-glucose (ICN).
Following incubation of neuronal cultures in an anaerobic chamber (Don Whitely
Scientific, England) for 35 minutes, the media from each well are removed and
replaced with 50 1 BSS and 50 l of Neurobasal/2% N2 supplement (Invitrogen)
before
placing culture wells into a CO2 incubator. Control neuronal cultures received
the
same BSS wash procedures and media additions as ischemic cultures, but were
maintained in a CO2 incubator. After a further 24 h, neuronal viability was
assessed
using the MTS assay as described in the preceding examples.
Results
The results from this assay indicated that the retroinverted PhylomerTM
peptides
PYC35D-TAT and PYC36D-TAT are protective (p <0.0001), when they were added
before or immediately after OGD (Figure 9).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
72
EXAMPLE 6
Peptides that inhibit AP-1 signaling have half-lives sufficient for therapy of
acute
ischemic events

Having established the biologically efficacy of the retro-inverso forms of the
neuroprotective PhylomerTM peptides, their in vitro stabilities were assessed.

Materials and methods
1. In vitro plasma stability
A stock solution of 200 g/ml of peptide in PBS was diluted into 1.5ml of human
plasma or PBS to achieve a final concentration of 10 g/ml. Of this solution,
50 1 was
then dispensed into 200 1 Max Recovery tubes (Axygen Scientific) in triplicate
for
each time point needed in the study. The tubes were incubated at 37 C and at
the
appropriate time point, they were removed from the incubator and 150 1 of
extraction
solution (75% acetonitrile/25% water + 2%formic acid) was added. The tubes
were
vortexed for 15 sec and then centrifuged at 11,000 RPM for 5mins at 4 C. Then,
100 l
of the supernatant was transferred into HPLC vials for analysis.

A fixed mobile phase was used so that all of the different charged species of
peptide
would elute at the same time, allowing a mass measurement of non-degraded
material.
Injection volume: 10 l; Column: Phenomenex Aqua C18 5 M 150mm X 4.6mm;
Flow: 0.8 ml/min; 85% Mobile phase A: 4% fomlic acid in nanopure water; 15%
Mobile phase B: acetonitrile; RT: 1.3 minutes; Stop time: 3:0 minutes. Mass
Spectrometer: MS-03 LC/MSD Trap XCT Ultra.
2. In vivo plasma stability/clearance rate
Male Sprague-Dawley rats (276-310g) were dosed with peptide by i.v injection
via the
jugular vein. The calculated dose for each animal was 3 mg/kg of body weight
and each
isoflurane anaesthetised animal received dosing volumes of 0.1m1/100g of body
weight of a 3mg/mi solution. The remaining dosing solutions were subjected to
a
similar protocol as the solutions that were administered to the animals and
were


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
73
collected pre-labeled Maxymum Recovery (Axygen Scientific) low binding tubes,
ready for quantification. Blood samples (0.2 ml) were collected just prior to
dosing
(pre-dose) and at various time points post-dosing according to the
experimental
schedule. Blood samples were collected from the jugular vein from the
anaesthetized
and transferred into 0.5m1 pre-labeled EDTA micro-centrifuge tubes and placed
on ice.
The tubes were then centrifu.ged for 5mins at 3,000 RPM in a bench top
microcentrifuge (4 C). The plasma was transferred to pre-labeled 1.7 ml tubes,
stored
in a-80 C freezer prior to analysis by LC-MS.

Analysis: Sample Preparation: 50 l of plasma was added to 150 .l of
precipitating
solvent (75% acetonitrile/25% water+2%formic acid) and vortexed for 15 sec.
The
solution was centrifuged at 11,000 rpm at 4 C for 5mins. 100 gl of the
supernatant was
transferred to a HPLC vial for quantification.

The HPLC conditions were as follows: Injection: 10 l; Column: Phenomenex Aqua
C18 5 M 150mm X 4.6mm; Flow: 0.7 ml/min; Mobile phase A: 4% formic acid in
nanopure water., Mobile phase B: acetonitrile., Retention time: 2.2mins., Mass
Spectrometer: MS-03 LC/MSD Trap XCT Ultra.

2o Results
An analysis of the full-length PhylomerTM peptides remaining in freshly drawn
human
plasma using a liquid chromatography/Mass Spectrometry (LC-MS) assay showed
that
PYC35D-TAT and PYC36D-TAT have half-lives of more than 12 hours (Figure 10).
To test in vivo clearance rates, these PhylomerTM peptides were injected into
rats and, at
various time points, blood samples were taken and analysed to quantify the
remaining
full-length peptide by LC-MS. Peptides PYC35D-TAT and PYC36D-TAT had in vivo
half-lives of 35 minutes and 100 minutes, respectively (data not shown).

These in vivo half-lives are in a range which would allow application in an
acute
treatment setting. This is appropriate for emergency therapy for stroke or
traumatic
brain injury, being comparable with that of tissue plasminogen activator
(tPA), the


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
74
leading emergency stroke therapy, which has an in vivo half life of a few
minutes in
rabbit blood. The biological efficacy of these peptides ex vivo and in vivo
establish
their suitability for acute therapeutic regimes, without necessarily the need
for prior
affinity maturation.

EXAMPLE 7
Peptides that inhibit AP-1 signaling are neuroprotective in a rat model of
global
cerebral ischemia

As PYC35D-TAT and PYC36D-TAT were active ex vivo in neuroprotection assays,
and exhibited long half-lives in vivo, their activities were tested in a rat
model of global
cerebral ischemia. For comparison, the JNK1-ID-TAT peptide was included as a
control.

Materials and methods
This study was approved by the Animal Ethics Committee of the University of
Western
Australia. The 2 vessel common carotid occlusion with hypotension model was
used to
induce global cerebral ischemia in 8-10 week old adult male Sprague-Dawley
rats
(Miles et al., 2001; Zhu et al., 2004). During the procedure, both cranial and
rectal
temperatures were measured via a thermocouple (Physitemp, New Jersey, USA),
and
were maintained at 37 0.2 C with a heating fan and pad. Rats were
anesthetized with
halothane/27% 02/balanced NO2 and ventilated before, during and for at least
15 min
after global cerebral ischemia. Cerebral ischemia was recorded from the time
the EEG
became iso-electric and was maintained for a duration of 8 minutes. Blood
pressure
was reduced by exsanguination to a maintenance level between 35 and 40 mmHg
during ischemia. Ten minutes before and 15 min after the ischemic insult,
Pa02,
PaCOz and pH were measured with a pH/blood gas analyzer (ABL5 Radiometer,
Copenhagen, Denmark).

Peptide doses of 11 nmoles were administered via injection to the cerebral
ventricle 1
hour post ischemia. Control animals consisted of sham-operated animals and
ischemic


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
animals treated with saline. Post-surgery animals were given 5m1 of warmed
0.9%
NaC1 by SC injection and placed in a clean cage where rectal temperature was
monitored and maintain between 37.0 and 38.0 C with heating/cooling fan as
required.

5 At 7 days post-ischemia animals were killed and CA1 neuronal survival was
assessed
by counting the number of normal-appearing pyramidal neurons per high-power
field
(400x) in 1000 gm segments in the medial, intermediate and lateral sections of
the
hippocampal CAl region (bregma section -3.8). CAl counts were expressed as a
percentage of sham values, which was taken as 100% neuronal survival. Neuronal
cell
10 counts were conducted by an observer who was blinded to the experimental
protocol.
Physiological parameters (blood pressure, gases, glucose, pH, cranial
temperature) and
CAl neuronal counts were analyzed by ANOVA. If significant variance was
observed
post hoc Bonferroni/Dunn pair wise comparisons were made. All data are
presented as
15 mean standard deviation. A value of P < 0.05 was considered significant.

Results
Whilst not optimised for dosage (dose chosen to match published dosage of the
D-JNKI
positive control), peptides PYC35D-TAT and PYC36D-TAT blocked neuronal cell
20 death in this model when injected intracerebroventricularly, demonstrating
their
efficacy in vivo (Figure 11).

EXAMPLE 8
Peptides that inhibit AP-1 signaling also inhibit neuronal apoptosis as
determined
25 by loss of MAP2 immunoreactivity

The ability of AP-1 signaling-inhibitory peptides to inhibit neuronal
apoptosis was
assessed by loss of MAP2 immunoreactivity in a focal model of traumatic brain
injury
(Chung et al., J. Neuroscience 23, 3336-3342, 2003; King et al., Neuropathol
Appl
3o Neurobiol. 27, 115-126, 2001).


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
76
Materials and fnethods
Rats were subjected to acute cortical injury as previously described by Chung
et al., J.
Neuroscience 23, 3336-3342 (2003) and King et al., Neuropathol Appl Neurobiol.
27,
115-126 (2001). Briefly, male 240g Hooded-Wistar rats were deeply
anaesthetized
using Isofluorane (2-3%) and were given an analgesic (Meloxycam, 40mg/kg), by
subcutaneous injection. Animals were immobilized in a Stoelting stereotactic
frame,
and a 21 gauge blunt Hamilton syringe was inserted into the Parl region of the
somatosensory cortex to a depth of 2mm and was left in place for 10 minutes.
Then,
0.5 gl of 5 M peptide was administered through the syringe at a rate of 0.05
l/min.
1o Peptide (l0 l) was applied to 1mm2 of Gelfoam, which was implanted before
suturing.
At 1 day post-injury (PI), rats were deeply anaesthetized with sodium
pentobarbital
(60mg/kg), then perfused transcardially with a 4% paraformalydehyde and 4%
sucrose
fixative solution in 0.O1M PBS. Brains were removed and cryoprotected in 18%
and
30% sucrose solutions, respectively and 40 m coronal sections were collected
serially
through the injury site for immunohistochemical labelling. All animal
procedures were
approved by the University of Tasmania Animal Ethics Committee (Permit
A008878).
Sections were labelled with Nissl Red, Nuclear Yellow and anti-MAP2 to allow a
quantitative assessment of neuronal loss around the lesion. Sections from the
lesion
centre and 200 m anterior and posterior to the centre of the injury, at layer
III of the
neocortex were included in the analysis and the area of MAP2 loss was scored
by
blinded observers. Three sections from each of three animals were examined for
each
treatment and significance assessed by ANOVA and post-hoc comparison (Tukey
test).

All data are presented as mean standard error. A value of P < 0.05 was
considered
significant.

Results
In this model, PYC35D-TAT was neuroprotective when compared with a negative
control peptide designated PYC35D Scram-TAT in which the PhylomerTm peptide
moiety was scrambled (p<0.05) (Figures 12,13). Peptide PYC35SD-TAT provided


CA 02663545 2009-03-13
WO 2008/034161 PCT/AU2007/000092
77
greater neuroprotection in this head injury model than the peptide JNK1-ID-TAT
(Figure 12).

Together, these studies established that these AP-1 signaling inhibitory
Phylomer'FM
peptides provide neuroprotective activity both in vitro and in vivo, and are
promising
leads for therapeutic development, especially for acute treatment settings
such as in
migraine, stroke, acute head trauma injury and organ reperfusion injury.

Representative Drawing

Sorry, the representative drawing for patent document number 2663545 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2007-01-31
(87) PCT Publication Date 2008-03-27
(85) National Entry 2009-03-13
Examination Requested 2012-01-18
Dead Application 2014-01-31

Abandonment History

Abandonment Date Reason Reinstatement Date
2013-01-31 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2009-03-13
Maintenance Fee - Application - New Act 2 2009-02-02 $100.00 2009-03-13
Maintenance Fee - Application - New Act 3 2010-02-01 $100.00 2009-03-13
Registration of a document - section 124 $100.00 2009-05-15
Maintenance Fee - Application - New Act 4 2011-01-31 $100.00 2010-12-10
Maintenance Fee - Application - New Act 5 2012-01-31 $200.00 2011-12-07
Request for Examination $800.00 2012-01-18
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
PHYLOGICA LIMITED
Past Owners on Record
FEAR, MARK
MILECH, NADIA
WATT, PAUL
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2009-03-13 1 58
Claims 2009-03-13 8 379
Description 2009-03-13 77 4,081
Drawings 2009-03-13 18 842
Cover Page 2009-08-04 2 36
Description 2009-03-14 94 4,447
Assignment 2009-06-12 1 43
Correspondence 2009-05-27 1 26
Assignment 2009-03-13 3 85
PCT 2009-03-13 3 140
Assignment 2009-05-15 5 152
Prosecution-Amendment 2009-03-13 19 419
Prosecution-Amendment 2012-01-18 2 77

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :